1
|
Yao J, Lin X, Zhang X, Xie M, Ma X, Bao X, Song J, Liang Y, Wang Q, Xue X. Predictive biomarkers for immune checkpoint inhibitors therapy in lung cancer. Hum Vaccin Immunother 2024; 20:2406063. [PMID: 39415535 PMCID: PMC11487980 DOI: 10.1080/21645515.2024.2406063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 09/05/2024] [Accepted: 09/15/2024] [Indexed: 10/18/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) have changed the treatment mode of lung cancer, extending the survival time of patients unprecedentedly. Once patients respond to ICIs, the median duration of response is usually longer than that achieved with cytotoxic or targeted drugs. Unfortunately, there is still a large proportion of lung cancer patients do not respond to ICI. Effective biomarkers are crucial for identifying lung cancer patients who can benefit from them. The first predictive biomarker is programmed death-ligand 1 (PD-L1), but its predictive value is limited to specific populations. With the development of single-cell sequencing and spatial imaging technologies, as well as the use of deep learning and artificial intelligence, the identification of predictive biomarkers has been greatly expanded. In this review, we will dissect the biomarkers used to predict ICIs efficacy in lung cancer from the tumor-immune microenvironment and host perspectives, and describe cutting-edge technologies to further identify biomarkers.
Collapse
Affiliation(s)
- Jie Yao
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xuwen Lin
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xin Zhang
- Department of Respiratory and Critical Care, Shandong Second Medical University, Weifang, Shandong, China
| | - Mei Xie
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xidong Ma
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xinyu Bao
- Department of Respiratory and Critical Care, Shandong Second Medical University, Weifang, Shandong, China
| | - Jialin Song
- Department of Respiratory and Critical Care, Shandong Second Medical University, Weifang, Shandong, China
| | - Yiran Liang
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Qiqi Wang
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xinying Xue
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Department of Respiratory and Critical Care, Shandong Second Medical University, Weifang, Shandong, China
| |
Collapse
|
2
|
Johnson SS, Liu D, Ewald JT, Robles-Planells C, Pulliam C, Christensen KA, Bayanbold K, Wels BR, Solst SR, O’Dorisio MS, Menda Y, Spitz DR, Fath MA. Auranofin inhibition of thioredoxin reductase sensitizes lung neuroendocrine tumor cells (NETs) and small cell lung cancer (SCLC) cells to sorafenib as well as inhibiting SCLC xenograft growth. Cancer Biol Ther 2024; 25:2382524. [PMID: 39054566 PMCID: PMC11275529 DOI: 10.1080/15384047.2024.2382524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/23/2024] [Accepted: 07/16/2024] [Indexed: 07/27/2024] Open
Abstract
Thioredoxin Reductase (TrxR) functions to recycle thioredoxin (Trx) during hydroperoxide metabolism mediated by peroxiredoxins and is currently being targeted using the FDA-approved anti-rheumatic drug, auranofin (AF), to selectively sensitize cancer cells to therapy. AF treatment decreased TrxR activity and clonogenic survival in small cell lung cancer (SCLC) cell lines (DMS273 and DMS53) as well as the H727 atypical lung carcinoid cell line. AF treatment also significantly sensitized DMS273 and H727 cell lines in vitro to sorafenib, an FDA-approved multi-kinase inhibitor that depleted intracellular glutathione (GSH). The pharmacokinetic, pharmacodynamic, and safety profile of AF was examined in nude mice with DMS273 xenografts administered AF intraperitoneally at 2 mg/kg or 4 mg/kg (IP) once (QD) or twice daily (BID) for 1-5 d. Plasma levels of AF were 10-20 μM (determined by mass spectrometry of gold), and the optimal inhibition of TrxR activity was obtained at 4 mg/kg once daily, with no effect on glutathione peroxidase 1 activity. This AF treatment extended for 14 d, inhibited TrxR (>75%), and resulted in a significant prolongation of median overall survival from 19 to 23 d (p = .04, N = 30 controls, 28 AF). In this experiment, there were no observed changes in animal bodyweight, complete blood counts (CBCs), bone marrow toxicity, blood urea nitrogen, or creatinine. These results support the hypothesis that AF effectively inhibits TrxR both in vitro and in vivo in SCLC, sensitizes NETs and SCLC to sorafenib, and could be repurposed as an adjuvant therapy with targeted agents that induce disruptions in thiol metabolism.
Collapse
Affiliation(s)
- Spenser S. Johnson
- Department of Radiation Oncology, Holden Comprehensive Cancer Center, Free Radical and Radiation Biology Program, University of Iowa Hospitals and Clinics, IA, USA
| | - Dijie Liu
- Department Pediatrics, University of Iowa Hospitals and Clinics, IA, USA
| | - Jordan T. Ewald
- Department of Radiation Oncology, Holden Comprehensive Cancer Center, Free Radical and Radiation Biology Program, University of Iowa Hospitals and Clinics, IA, USA
| | | | - Casey Pulliam
- Department of Radiation Oncology, Holden Comprehensive Cancer Center, Free Radical and Radiation Biology Program, University of Iowa Hospitals and Clinics, IA, USA
| | - Keegan A. Christensen
- Department of Radiation Oncology, Holden Comprehensive Cancer Center, Free Radical and Radiation Biology Program, University of Iowa Hospitals and Clinics, IA, USA
| | - Khaliunaa Bayanbold
- Department of Radiation Oncology, Holden Comprehensive Cancer Center, Free Radical and Radiation Biology Program, University of Iowa Hospitals and Clinics, IA, USA
| | - Brian R. Wels
- State Hygienic Laboratory, University of Iowa, IA, USA
| | - Shane R. Solst
- Department of Radiation Oncology, Holden Comprehensive Cancer Center, Free Radical and Radiation Biology Program, University of Iowa Hospitals and Clinics, IA, USA
| | - M. Sue O’Dorisio
- Department Pediatrics, University of Iowa Hospitals and Clinics, IA, USA
| | - Yusuf Menda
- Department of Radiology, Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, IA, USA
| | - Douglas R. Spitz
- Department of Radiation Oncology, Holden Comprehensive Cancer Center, Free Radical and Radiation Biology Program, University of Iowa Hospitals and Clinics, IA, USA
| | - Melissa A. Fath
- Department of Radiation Oncology, Holden Comprehensive Cancer Center, Free Radical and Radiation Biology Program, University of Iowa Hospitals and Clinics, IA, USA
| |
Collapse
|
3
|
Jiang Y, Xie J, Cheng Q, Cai Z, Xu K, Lu W, Wang F, Wu X, Song Y, Lv T, Zhan P. Comprehensive genomic and spatial immune infiltration analysis of survival outliers in extensive-stage small cell lung cancer receiving first-line chemoimmunotherapy. Int Immunopharmacol 2024; 141:112901. [PMID: 39151386 DOI: 10.1016/j.intimp.2024.112901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/20/2024] [Accepted: 08/05/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND A minority of patients with extensive-stage small cell lung cancer (ES-SCLC) exhibit prolonged survival following first-line chemoimmunotherapy, which warrants the use of reliable biomarkers. Here, we investigated the disparities in genomics and immune cell spatial distribution between long- and short-term survival of patients with ES-SCLC. METHODS We retrospectively recruited 11 long-term (>2 years) and 13 short-term (<9 months) ES-SCLC survivors receiving first-line chemoimmunotherapy. The samples were processed using targeted next-generation sequencing (tNGS), programmed death ligand-1 staining, multiplex immunohistochemical staining for immune cells (mIHC), tumor mutation burden (TMB), and chromosomal instability score measurements. The expression of putative genes in SCLC at the bulk and single-cell RNA-sequencing levels, as well as the role of putative genes in pan-cancer immunotherapy cohorts, were analyzed. RESULTS At the genomic level, a greater proportion of the smoking signature and higher TMB (>3.1) were associated with favorable survival. At the single-gene and pathway levels, tNGS revealed that MCL1 and STMN1 amplification and alterations in the apoptosis pathway were more common in short-term survivors, whereas alterations in the DLL3, KMT2B, HGF, EPHA3, ADGRB3, lysine deprivation, and HGF-cMET pathways were observed more frequently in long-term survivors. mIHC analysis of immune cells with different spatial distributions revealed that long-term survivors presented increased numbers of M1-like macrophages in all locations and decreased numbers of CD8+ T cells in the tumor stroma. Bulk transcriptomic analysis demonstrated that high levels of STMN1 and DLL3 represented an immune-suppressive tumor immune microenvironment (TIME), whereas HGF indicated an immune-responsive TIME. The expression levels of our putative genes were comparative in both TP53/RB1 mutant-type and TP53/RB1 wild-type. At the single-cell level, STMN1, MCL1, and DLL3 were highly expressed among all molecular subtypes (SCLC-A, SCLC-N, and SCLC-P), with STMN1 being enriched in cell division and G2M checkpoint pathways. CONCLUSIONS For ES-SCLC patients receiving first-line chemoimmunotherapy, alterations in DLL3, KMT2B, HGF, EPHA3, and ADGRB3 and a greater proportion of M1-like macrophages infiltration in all locations were predictors of favorable survival, while MCL1 and STMN1 amplification, as well as a greater proportion of CD8+ T cells infiltrating the tumor stroma, predicted worse survival.
Collapse
Affiliation(s)
- Yuxin Jiang
- School of Medicine, Southeast University, Nanjing 210000, China
| | - Jingyuan Xie
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China
| | - Qinpei Cheng
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China
| | - Zijing Cai
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Nanjing Medical School, Nanjing 210002, China
| | - Ke Xu
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China
| | - Wanjun Lu
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China
| | - Fufeng Wang
- Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, China
| | - Xiaoying Wu
- Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, China
| | - Yong Song
- School of Medicine, Southeast University, Nanjing 210000, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Nanjing Medical School, Nanjing 210002, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, School of Medicine, Southeast University, Nanjing 210002, China.
| | - Tangfeng Lv
- School of Medicine, Southeast University, Nanjing 210000, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Nanjing Medical School, Nanjing 210002, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, School of Medicine, Southeast University, Nanjing 210002, China.
| | - Ping Zhan
- School of Medicine, Southeast University, Nanjing 210000, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Nanjing Medical School, Nanjing 210002, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, School of Medicine, Southeast University, Nanjing 210002, China.
| |
Collapse
|
4
|
Seo Y, Zhang S, Jang J, Ko KP, Kim KB, Huang Y, Kim DW, Kim B, Zou G, Zhang J, Jun S, Chu W, Kirk NA, Hwang YE, Ban YH, Dhar SS, Chan JM, Lee M, Rudin CM, Park KS, Park JI. Actin dysregulation induces neuroendocrine plasticity and immune evasion: a vulnerability of small cell lung cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.02.15.528365. [PMID: 36824957 PMCID: PMC9949038 DOI: 10.1101/2023.02.15.528365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
Small cell lung cancer (SCLC) is aggressive with limited therapeutic options. Despite recent advances in targeted therapies and immunotherapies, therapy resistance is a recurring issue, which might be partly due to tumor cell plasticity, a change in cell fate. Nonetheless, the mechanisms underlying tumor cell plasticity and immune evasion in SCLC remain elusive. CRACD, a capping protein inhibitor that promotes actin polymerization, is frequently inactivated in SCLC. Cracd knockout (KO) transforms preneoplastic cells into SCLC tumor-like cells and promotes in vivo SCLC development driven by Rb1, Trp53, and Rbl2 triple KO. Cracd KO induces neuroendocrine (NE) plasticity and increases tumor cell heterogeneity of SCLC tumor cells via dysregulated NOTCH1 signaling by actin cytoskeleton disruption. CRACD depletion also reduces nuclear actin and induces EZH2-mediated H3K27 methylation. This nuclear event suppresses the MHC-I genes and thereby depletes intratumoral CD8+ T cells for accelerated SCLC tumorigenesis. Pharmacological blockade of EZH2 inhibits CRACD-negative SCLC tumorigenesis by restoring MHC-I expression and immune surveillance. Unsupervised single-cell transcriptomics identifies SCLC patient tumors with concomitant inactivation of CRACD and downregulated MHC-I pathway. This study defines CRACD, an actin regulator, as a tumor suppressor that limits cell plasticity and immune evasion and proposes EZH2 blockade as a viable therapeutic.
Collapse
|
5
|
Varlotto J, Voland R, DeCamp M, Khatri J, Shweihat Y, Nwanwene K, Tirona M, Wright T, Pacioles T, Jamil M, Anwar K, Bastidas J, Chowdhury N, Zander D, Silbermins D, Abdallah M, Flickinger J. Role of consolidative thoracic and prophylactic cranial radiation in extensive stage small cell lung cancer in chemo-immunotherapy era. Radiother Oncol 2024; 202:110619. [PMID: 39537032 DOI: 10.1016/j.radonc.2024.110619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/28/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
INTRODUCTION The role of consolidative thoracic and prophylactic brain radiation for extensive stage small cell lung cancer patients is controversial. We investigated the factors associated with the use of any radiation therapy (RT) and whether RT has a benefit to overall survival (OS) in patients receiving any systemic therapy and whether this benefit is the same if Chemotherapy (CT) or chemo-immunotherapy (CT-IO) is used. MATERIAL/METHODS The NCDB database was queried from years 2017-2019. Patients receiving systemic therapy- STX (CT or CT-IO) had to have at least 6 months of follow-up and have no brain metastases at diagnosis. All RT patients had to receive upfront systemic therapy, be treated 2-6 months from diagnosis, and if treated to the brain received 25 Gy in 10 fractions only. Multi-variable analyses (MVA) were used to determine factors associated with OS and selection for any radiation. Propensity matching for factors affecting OS were used to generate Kaplan-Meier OS curves. Log-rank tests were used to determine differences in Kaplan Meier survival curves for the effects of RT on OS. RESULTS The total number of patients receiving RT/STX or STX alone as well as their median follow-up (months) were (890, 17.0 mn) and (6898, 14.0mn). The median time to the start of STX and RT were 22.9 days and 152 days, respectively. MVA noted that RT had a greater effect on OS (Thorax, Brain, Both Brain/Thorax - HRs = 0.80, 0.77, 0.70) than other interventions including IO (HR 0.87) and palliative care without RT (HR 1.06). Selection for radiation depended significantly upon factors affecting OS (HR) including lack of liver metastases, females, age and Charlson co-morbidity index, but did not depend upon insurance status, race, or county income/high school graduation rates. Propensity-score matched OS curves noted the same significant effects of RT on OS in those receiving CT +/- IO, CT-IO, and CT alone with HRs of 0.68/0.68/0.68 for thoracic RT, 0.72/0.72/0.70 for brain RT, and 0.60/0.60/0.60 for brain/thoracic RT, respectively. CONCLUSIONS The patient with extensive stage small cell lung cancer who reach candidacy and receive RT may have a significant improvement in OS compared to the patients treated only with CT or CT-IO. Combined thoracic and prophylactic brain RT seems to be better than either one alone. The impact of radiation whether given to one or two sites may be more beneficial than immunotherapy added to chemotherapy.
Collapse
Affiliation(s)
- J Varlotto
- Department of Oncology, Edwards Cancer Institute/Marshall University, Huntington, WV, United States.
| | - R Voland
- Department of Ophthalmology, University of Wisconsin, Madison, WI, United States
| | - M DeCamp
- Division of Thoracic Surgery, University of Wisconsin, Madison, WI, United States
| | - J Khatri
- Department of Oncology, Edwards Cancer Institute/Marshall University, Huntington, WV, United States
| | - Y Shweihat
- Department of Internal Medicine, Marshall Health, Huntington, WV, United States
| | - K Nwanwene
- Department of Oncology, Edwards Cancer Institute/Marshall University, Huntington, WV, United States
| | - M Tirona
- Department of Oncology, Edwards Cancer Institute/Marshall University, Huntington, WV, United States
| | - T Wright
- Department of Internal Medicine, Marshall Health, Huntington, WV, United States
| | - T Pacioles
- Department of Oncology, Edwards Cancer Institute/Marshall University, Huntington, WV, United States
| | - M Jamil
- Department of Oncology, Edwards Cancer Institute/Marshall University, Huntington, WV, United States
| | - K Anwar
- Department of Oncology, Edwards Cancer Institute/Marshall University, Huntington, WV, United States
| | - J Bastidas
- Department of Surgery, Marshall University, Huntington, WV, United States
| | - N Chowdhury
- Department of Surgery, Marshall University, Huntington, WV, United States
| | - D Zander
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - D Silbermins
- Department of Oncology, Edwards Cancer Institute/Marshall University, Huntington, WV, United States
| | - M Abdallah
- Department of Oncology, Edwards Cancer Institute/Marshall University, Huntington, WV, United States
| | - J Flickinger
- Department of Radiation Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| |
Collapse
|
6
|
Ren Z, Shang S, Chen D. Recent advances in immunotherapy for small cell lung cancer. Curr Opin Oncol 2024:00001622-990000000-00220. [PMID: 39526685 DOI: 10.1097/cco.0000000000001105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
PURPOSE OF REVIEW This review aims to provide an overview of recent advances in immunotherapy for small cell lung cancer (SCLC), with a focus on the current status of immune checkpoint inhibitors (ICIs), novel combination strategies, and key biomarkers. RECENT FINDINGS The integration of ICIs into standard chemotherapy has established them as the first-line treatment for extensive-stage SCLC (ES-SCLC). The ADRIATIC trial further demonstrated the efficacy of ICI maintenance therapy in limited-stage SCLC. Additionally, combining radiotherapy with ICIs has shown promising synergistic effects, including the abscopal and radscopal effects. Ongoing investigations into the combination of ICIs with targeted therapies, such as antiangiogenic agents and DNA damage response inhibitors, have yielded encouraging preliminary results. Notably, the novel therapeutic agent tarlatamab, the first bispecific DLL3-directed CD3 T-cell engager, has recently received FDA approval for second-line treatment of ES-SCLC. Advances in omics technologies have shed light on the intra-tumor and inter-tumor heterogeneity of SCLC, leading to the identification of new molecular subtypes and biomarkers, thereby paving the way for precision medicine. SUMMARY Despite the improved outcomes associated with immunotherapy in SCLC, the overall clinical benefit remains modest. Further preclinical and clinical studies are essential to identify optimal treatment regimens and enhance therapeutic efficacy.
Collapse
Affiliation(s)
- Ziyuan Ren
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | | | | |
Collapse
|
7
|
Rodarte KE, Heyman SN, Guo L, Flores L, Savage TK, Villarreal J, Deng S, Xu L, Shah RB, Oliver TG, Johnson JE. Neuroendocrine Differentiation in Prostate Cancer Requires ASCL1. Cancer Res 2024; 84:3522-3537. [PMID: 39264686 PMCID: PMC11534540 DOI: 10.1158/0008-5472.can-24-1388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/04/2024] [Accepted: 09/06/2024] [Indexed: 09/13/2024]
Abstract
Most patients with prostate adenocarcinoma develop resistance to therapies targeting the androgen receptor (AR). Consequently, a portion of these patients develop AR-independent neuroendocrine (NE) prostate cancer (NEPC), a rapidly progressing cancer with limited therapies and poor survival outcomes. Current research to understand the progression to NEPC suggests a model of lineage plasticity whereby AR-dependent luminal-like tumors progress toward an AR-independent NEPC state. Genetic analysis of human NEPC identified frequent loss of RB1 and TP53, and the loss of both genes in experimental models mediates the transition to a NE lineage. Transcriptomics studies have shown that lineage transcription factors ASCL1 and NEUROD1 are present in NEPC. In this study, we modeled the progression of prostate adenocarcinoma to NEPC by establishing prostate organoids and subsequently generating subcutaneous allograft tumors from genetically engineered mouse models harboring Cre-induced loss of Rb1 and Trp53 with Myc overexpression (RPM). These tumors were heterogeneous and displayed adenocarcinoma, squamous, and NE features. ASCL1 and NEUROD1 were expressed within NE-defined regions, with ASCL1 being predominant. Genetic loss of Ascl1 in this model did not decrease tumor incidence, growth, or metastasis; however, there was a notable decrease in NE identity and an increase in basal-like identity. This study provides an in vivo model to study progression to NEPC and establishes the requirement for ASCL1 in driving NE differentiation in prostate cancer. Significance: Modeling lineage transitions in prostate cancer and testing dependencies of lineage transcription factors have therapeutic implications, given the emergence of treatment-resistant, aggressive forms of neuroendocrine prostate cancer. See related commentary by McQuillen and Brady, p. 3499.
Collapse
Affiliation(s)
- Kathia E. Rodarte
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Shaked Nir Heyman
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Lei Guo
- Quantitative Biomedical Research Center, Peter O’Donnell Jr. School of Public Health, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Lydia Flores
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Trisha K. Savage
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Juan Villarreal
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Su Deng
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Lin Xu
- Quantitative Biomedical Research Center, Peter O’Donnell Jr. School of Public Health, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Pediatrics, Division of Hematology/Oncology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Rajal B. Shah
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Trudy G. Oliver
- Department of Pharmacology and Cancer Biology, Duke University, NC 27708, USA
| | - Jane E. Johnson
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
8
|
Lucia F, Antoni D, Vaugier L, Duvergé L, Thureau S, Bourbonne V. Role of stereotactic radiotherapy in the management of small-cell lung cancer. Cancer Radiother 2024; 28:628-632. [PMID: 39358196 DOI: 10.1016/j.canrad.2024.07.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 10/04/2024]
Abstract
Small-cell lung cancer is the most aggressive form of lung neoplasia, treated in recent decades with chemoradiotherapy in case of limited stage and chemotherapy alone at the metastatic stage. In the last few years, the advent of immunotherapy has changed the landscape in the treatment of non-small-cell lung cancer, and to a lesser degree in small-cell lung cancer. Despite the recent advances in research, small-cell lung cancer is still considered an aggressive and lethal disease characterized by high recurrence or metastatic potential. As stereotactic radiotherapy has established itself as the standard of care in the early stage of inoperable non-small-cell lung cancer and in metastatic disease to treat brain and extracranial metastases, these same issues now arise in the management of small-cell lung cancer. This article aims to review the current knowledge and the potential of stereotactic radiotherapy in small-cell lung cancer.
Collapse
Affiliation(s)
- François Lucia
- Radiation Oncology Department, CHU de Brest, 2, avenue Foch, 29609 Brest cedex, France; LaTIM, UMR 1101, Inserm, université de Bretagne occidentale, Brest, France.
| | - Delphine Antoni
- Radiation Oncology Department, institut de cancérologie Strasbourg Europe, Strasbourg, France
| | - Loïg Vaugier
- Radiation Oncology Department, institut de cancérologie de l'Ouest - centre René-Gauducheau, boulevard Jacques-Monod, 44800 Saint-Herblain, France
| | - Loïg Duvergé
- Radiation Oncology Department, centre Eugène-Marquis, avenue de la Bataille Flandres-Dunkerque, 35000 Rennes, France
| | - Sébastien Thureau
- Radiation Oncology Department, centre Henri-Becquerel, Rouen, France; QuantIf-Litis EA4108, université de Rouen, Rouen, France
| | - Vincent Bourbonne
- Radiation Oncology Department, CHU de Brest, 2, avenue Foch, 29609 Brest cedex, France; LaTIM, UMR 1101, Inserm, université de Bretagne occidentale, Brest, France
| |
Collapse
|
9
|
Velut Y, Arqué B, Wislez M, Blons H, Burroni B, Prieto M, Beau S, Fournel L, Birsen G, Cremer I, Alifano M, Damotte D, Mansuet-Lupo A. The tumor immune microenvironment of SCLC is not associated with its molecular subtypes. Eur J Cancer 2024; 212:115067. [PMID: 39413714 DOI: 10.1016/j.ejca.2024.115067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 08/28/2024] [Accepted: 10/10/2024] [Indexed: 10/18/2024]
Abstract
INTRODUCTION Small-cell lung carcinoma (SCLC) is a high-grade neuroendocrine carcinoma of poor prognosis. Although immune checkpoint blockers have shown promising results in advanced SCLC, the tumor immune microenvironment (TME) remains poorly understood, with no validated prognostic or predictive biomarkers of efficacy. METHODS This retrospective study included surgically samples from 48 SCLC patients between 2009 and 2018. We assessed the TME using two quantitative 7-plex immunofluorescence panels focusing on T and B cells, and compared it to NSCLC (N = 10). Molecular subtypes were determined by assessing the expression of ASCL1, NEUROD1 and YAP1 using immunohistochemistry. RESULTS Immune-hot SCLC were defined as those exhibiting the highest immune cell and immune-related marker densities. They were associated with longer overall survival, significantly more frequently detected at early stages, and exhibited high PD-L1 expression in immune cells, but were not associated with molecular subtypes. Compared to NSCLC, SCLC had significantly lower densities of CD20 + cells and higher density of PD1 + cells, with no significant differences in CD4 + , CD8 + and plasma cell densities. In univariate analysis, the highest OS was significantly associated with early stage (p < 0.001), low expression of NEUROD1 (p = 0.047), high PD1 + cell density (p < 0.001) and high PD-L1 immune cell expression (p = 0.04). Only stage and PD1 + cell density emerged as independent prognostic markers. CONCLUSION SCLC TME is highly heterogeneous. Immune-hot tumors were associated with OS but not with molecular classification. PD1 expression and PD-L1 expression by immune cells may thus serve as a prognostic marker.
Collapse
Affiliation(s)
- Yoan Velut
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, Institut du cancer Paris CARPEM, Team Inflammation, Complement and Cancer, Paris, France
| | - Basilia Arqué
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, Institut du cancer Paris CARPEM, Team Inflammation, Complement and Cancer, Paris, France
| | - Marie Wislez
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, Institut du cancer Paris CARPEM, Team Inflammation, Complement and Cancer, Paris, France; Department of pneumology, Cochin Hospital, AP-HP.centre, Université Paris Cité, Paris, France
| | - Hélène Blons
- Centre de Recherche des Cordeliers, INSERM CNRS SNC 5096, Sorbonne Université, Université Paris Cité, Institut du cancer Paris CARPEM, Paris, France; Department of Biochemistry, Unit of Pharmacogenetic and Molecular Oncology, Georges Pompidou European Hospital, AP-HP.centre, Université Paris Cité, Paris, France
| | - Barbara Burroni
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, Institut du cancer Paris CARPEM, Team Inflammation, Complement and Cancer, Paris, France; Department of Pathology, Cochin Hospital, AP-HP.centre, Université Paris Cité, Paris, France
| | - Mathilde Prieto
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, Institut du cancer Paris CARPEM, Team Inflammation, Complement and Cancer, Paris, France; Department of Thoracic Surgery, Cochin Hospital, AP-HP.centre, Université Paris Cité, Paris, France
| | - Siméon Beau
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, Institut du cancer Paris CARPEM, Team Inflammation, Complement and Cancer, Paris, France
| | - Ludovic Fournel
- Department of Thoracic Surgery, Cochin Hospital, AP-HP.centre, Université Paris Cité, Paris, France
| | - Gary Birsen
- Department of pneumology, Cochin Hospital, AP-HP.centre, Université Paris Cité, Paris, France
| | - Isabelle Cremer
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, Institut du cancer Paris CARPEM, Team Inflammation, Complement and Cancer, Paris, France
| | - Marco Alifano
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, Institut du cancer Paris CARPEM, Team Inflammation, Complement and Cancer, Paris, France; Department of Thoracic Surgery, Cochin Hospital, AP-HP.centre, Université Paris Cité, Paris, France
| | - Diane Damotte
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, Institut du cancer Paris CARPEM, Team Inflammation, Complement and Cancer, Paris, France; Department of Pathology, Cochin Hospital, AP-HP.centre, Université Paris Cité, Paris, France.
| | - Audrey Mansuet-Lupo
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, Institut du cancer Paris CARPEM, Team Inflammation, Complement and Cancer, Paris, France; Department of Pathology, Cochin Hospital, AP-HP.centre, Université Paris Cité, Paris, France
| |
Collapse
|
10
|
Concannon KF, Glisson BS, Doebele RC, Huang C, Marotti M, Camidge DR, Heymach JV. A Phase I Open-Label Study of Cediranib Plus Etoposide and Cisplatin as First-Line Therapy for Patients With Extensive-Stage Small-Cell Lung Cancer or Metastatic Neuroendocrine Non-Small-Cell Lung Cancer. Clin Lung Cancer 2024; 25:601-611. [PMID: 39307607 DOI: 10.1016/j.cllc.2024.08.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 08/27/2024] [Indexed: 11/05/2024]
Abstract
INTRODUCTION Small cell lung cancer (SCLC) is known to express high levels of the proangiogenic factor vascular endothelial growth factor (VEGF). We assessed the safety and tolerability of cediranib, an oral inhibitor of VEGF receptor tyrosine kinases, in combination with etoposide and cisplatin as first-line therapy for extensive-stage (ES) SCLC or metastatic lung neuroendocrine cancer (NEC). METHODS Patients received up to six 21-day cycles of etoposide (100 mg/m2, days 1-3) and cisplatin (80 mg/m2, day 1) with once-daily cediranib until disease progression or unacceptable toxicity. Cediranib dosing started at 30 mg with de-escalation cohorts planned based on cycle 1 dose-limiting toxicities (DLTs). An expansion cohort of 12 patients was enrolled at the recommended phase II dose. RESULTS Twenty-two patients (18 with ES SCLC, 4 with NEC) received treatment. Only 4 patients were enrolled at the 30 mg cediranib dose before other studies established 20 mg/day as the recommended dose with chemotherapy. Among the 18 patients enrolled at the 20-mg dose, common adverse events included nausea/vomiting, neutropenia, and diarrhea; 8 patients (44%) had grade 1 or 2 hypertension, and 2 (11%) had grade 3 hemoptysis. For all 18 patients, the objective response rate and median progression-free survival duration were 67% and 7.9 months. Plasma levels of VEGF were significantly higher, and those of soluble VEGFR2 were significantly lower, on day 22 than at baseline but were not correlated with tumor shrinkage. CONCLUSIONS Cediranib (20 mg) plus etoposide and cisplatin is well tolerated and has promising clinical activity.
Collapse
Affiliation(s)
- Kyle F Concannon
- Department of Hematology/Onscology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Bonnie S Glisson
- Department of Thoracic Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Robert C Doebele
- Division of Medical Oncology, Department of Medicine, University of Colorado Cancer Center, Aurora, CO
| | - Chao Huang
- Department of Medical Oncology, University of Kansas, Kansas City, KS
| | | | - D Ross Camidge
- Division of Medical Oncology, Department of Medicine, University of Colorado Cancer Center, Aurora, CO
| | - John V Heymach
- Department of Thoracic Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX; AstraZeneca, Macclesfield, United Kingdom.
| |
Collapse
|
11
|
Miglietta G, Russo M, Capranico G, Marinello J. Stimulation of cGAS-STING pathway as a challenge in the treatment of small cell lung cancer: a feasible strategy? Br J Cancer 2024; 131:1567-1575. [PMID: 39215193 PMCID: PMC11555062 DOI: 10.1038/s41416-024-02821-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 08/03/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024] Open
Abstract
Lung cancer has a significant incidence among the population and, unfortunately, has an unfavourable prognosis in most cases. The World Health Organization (WHO) classifies lung tumours into two subtypes based on their phenotype: the Non-Small Cell Lung Cancer (NSCLC) and the Small Cell Lung Cancer (SCLC). SCLC treatment, despite advances in chemotherapy and radiotherapy, is often unsuccessful for cancer recurrence highlighting the need to develop novel therapeutic strategies. In this review, we describe the genetic landscape and tumour microenvironment that characterize the pathological processes of SCLC and how they are responsible for tumour immune evasion. The immunosuppressive mechanisms engaged in SCLC are critical factors to understand the failure of immunotherapy in SCLC and, conversely, suggest that new signalling pathways, such as cGAS/STING, should be investigated as possible targets to stimulate an innate immune response in this subtype of lung cancer. The full comprehension of the innate immunity of cancer cells is thus crucial to open new challenges for successful immunotherapy in treating SCLC and improving patient outcomes.
Collapse
Affiliation(s)
- Giulia Miglietta
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Marco Russo
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Giovanni Capranico
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Bologna, Italy.
| | - Jessica Marinello
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Bologna, Italy.
| |
Collapse
|
12
|
Li L, Sun Y. Circulating tumor DNA methylation detection as biomarker and its application in tumor liquid biopsy: advances and challenges. MedComm (Beijing) 2024; 5:e766. [PMID: 39525954 PMCID: PMC11550092 DOI: 10.1002/mco2.766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 11/16/2024] Open
Abstract
Circulating tumor DNA (ctDNA) methylation, an innovative liquid biopsy biomarker, has emerged as a promising tool in early cancer diagnosis, monitoring, and prognosis prediction. As a noninvasive approach, liquid biopsy overcomes the limitations of traditional tissue biopsy. Among various biomarkers, ctDNA methylation has garnered significant attention due to its high specificity and early detection capability across diverse cancer types. Despite its immense potential, the clinical application of ctDNA methylation faces substantial challenges pertaining to sensitivity, specificity, and standardization. In this review, we begin by introducing the basic biology and common detection techniques of ctDNA methylation. We then explore recent advancements and the challenges faced in the clinical application of ctDNA methylation in liquid biopsies. This includes progress in early screening and diagnosis, identification of clinical molecular subtypes, monitoring of recurrence and minimal residual disease (MRD), prediction of treatment response and prognosis, assessment of tumor burden, and determination of tissue origin. Finally, we discuss the future perspectives and challenges of ctDNA methylation detection in clinical applications. This comprehensive overview underscores the vital role of ctDNA methylation in enhancing cancer diagnostic accuracy, personalizing treatments, and effectively monitoring disease progression, providing valuable insights for future research and clinical practice.
Collapse
Affiliation(s)
- Lingyu Li
- Central Laboratory & Shenzhen Key Laboratory of Epigenetics and Precision Medicine for CancersNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeShenzhenChina
| | - Yingli Sun
- Central Laboratory & Shenzhen Key Laboratory of Epigenetics and Precision Medicine for CancersNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeShenzhenChina
| |
Collapse
|
13
|
Ozakinci H, Alontaga AY, Cano P, Koomen JM, Perez BA, Beg AA, Chiappori AA, Haura EB, Boyle TA. Unveiling the Molecular Features of SCLC With a Clinical RNA Expression Panel. JTO Clin Res Rep 2024; 5:100723. [PMID: 39386315 PMCID: PMC11459576 DOI: 10.1016/j.jtocrr.2024.100723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 07/26/2024] [Accepted: 08/23/2024] [Indexed: 10/12/2024] Open
Abstract
Introduction The translation of gene expression profiles of SCLC to clinical testing remains relatively unexplored. In this study, gene expression variations in SCLC were evaluated to identify potential biomarkers. Methods RNA expression profiling was performed on 44 tumor samples from 35 patients diagnosed with SCLC using the clinically validated RNA Salah Targeted Expression Panel (RNA STEP). RNA sequencing (RNA-Seq) and immunohistochemistry were performed on two different SCLC cohorts, and correlation analyses were performed for the ASCL1, NEUROD1, POU2F3, and YAP1 genes and their corresponding proteins. RNA STEP and RNA-Seq results were evaluated for gene expression profiles and heterogeneity between SCLC primary and metastatic sites. RNA STEP gene expression profiles of independent SCLC samples (n = 35) were compared with lung adenocarcinoma (n = 160) and squamous cell carcinoma results (n = 25). Results The RNA STEP results were highly correlated with RNA-Seq and immunohistochemistry results. The dominant transcription regulator by RNA STEP was ASCL1 in 74.2% of the samples, NEUROD1 in 20%, and POU2F3 in 2.9%. The ASCL1, NEUROD1, and POU2F3 gene expression profiles were heterogeneous between primary and metastatic sites. SCLCs displayed markedly high expression for targetable genes DLL3, EZH2, TERT, and RET. SCLCs were found to have relatively colder immune profiles than lung adenocarcinomas and squamous cell carcinomas, characterized by lower expression of HLA genes, immune cell, and immune checkpoint genes, except the LAG3 gene. Conclusions Clinical-grade SCLC RNA expression profiling has value for SCLC subtyping, design of clinical trials, and identification of patients for trials and potential targeted therapy.
Collapse
Affiliation(s)
- Hilal Ozakinci
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Aileen Y. Alontaga
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Pedro Cano
- Department of Pathology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - John M. Koomen
- Department of Pathology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Bradford A. Perez
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
- Florida Cancer Specialists & Research Institute, Trinity Cancer Center, Trinity, Florida
| | - Amer A. Beg
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Alberto A. Chiappori
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Eric B. Haura
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Theresa A. Boyle
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
- Department of Pathology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| |
Collapse
|
14
|
Roskoski R. Targeted and cytotoxic inhibitors used in the treatment of lung cancers. Pharmacol Res 2024; 209:107465. [PMID: 39426470 DOI: 10.1016/j.phrs.2024.107465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 10/11/2024] [Indexed: 10/21/2024]
Abstract
Lung cancer is the leading cause of cancer deaths in the United States and the world. It is divided into two major types: small cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC). In the tumor-node-metastasis (TNM) cancer-staging classification system (Stages I/II/III/IV), the severity of neoplastic growth is characterized by the size of the tumor (T1 to T4), the extent of lymph node involvement (N0 to N3), and whether (M1) or not (M0) distant metastasis has occurred. Surgery is the treatment of choice for medically fit patients with Stage I/II NSCLC. Combination chemoradiotherapy and immune checkpoint inhibitor therapy are used across all NSCLC types. Oncogene-addicted tumors with sensitizing EGFR or BRAF mutations or activating ALK, ROS1 or NTRK translocations are treated with their cognate orally active small molecule protein kinase blockers. On the order of 20 % of NSCLCs bear activating mutations in EGFR and are treated with osimertinib and other kinase antagonists. SCLC, which accounts for approximately 15 % of lung cancer cases, is a deadly high-grade neuroendocrine carcinoma with a poor prognosis. Limited-stage SCLC is confined to one hemi-thorax and one radiation port and extensive-stage disease signifies those cancers that do not meet the criteria for limited-stage disease. Local treatment options to control thoracic disease include radiotherapy and surgery. In patients with extensive-stage disease, a platinum agent (cisplatin or carboplatin) combined with etoposide and an anti-PDL1 inhibitor (atezolizumab or durvalumab) for four cycles followed by anti-PDL1 maintenance therapy is the recommended first-line regimen.
Collapse
Affiliation(s)
- Robert Roskoski
- Blue Ridge Institute for Medical Research, 221 Haywood Knolls Drive, Hendersonville, NC 28791, United States.
| |
Collapse
|
15
|
Iglesia MD, Jayasinghe RG, Chen S, Terekhanova NV, Herndon JM, Storrs E, Karpova A, Zhou DC, Naser Al Deen N, Shinkle AT, Lu RJH, Caravan W, Houston A, Zhao Y, Sato K, Lal P, Street C, Martins Rodrigues F, Southard-Smith AN, Targino da Costa ALN, Zhu H, Mo CK, Crowson L, Fulton RS, Wyczalkowski MA, Fronick CC, Fulton LA, Sun H, Davies SR, Appelbaum EL, Chasnoff SE, Carmody M, Brooks C, Liu R, Wendl MC, Oh C, Bender D, Cruchaga C, Harari O, Bredemeyer A, Lavine K, Bose R, Margenthaler J, Held JM, Achilefu S, Ademuyiwa F, Aft R, Ma C, Colditz GA, Ju T, Oh ST, Fitzpatrick J, Hwang ES, Shoghi KI, Chheda MG, Veis DJ, Chen F, Fields RC, Gillanders WE, Ding L. Differential chromatin accessibility and transcriptional dynamics define breast cancer subtypes and their lineages. NATURE CANCER 2024:10.1038/s43018-024-00773-6. [PMID: 39478117 DOI: 10.1038/s43018-024-00773-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 04/24/2024] [Indexed: 11/06/2024]
Abstract
Breast cancer (BC) is defined by distinct molecular subtypes with different cells of origin. The transcriptional networks that characterize the subtype-specific tumor-normal lineages are not established. In this work, we applied bulk, single-cell and single-nucleus multi-omic techniques as well as spatial transcriptomics and multiplex imaging on 61 samples from 37 patients with BC to show characteristic links in gene expression and chromatin accessibility between BC subtypes and their putative cells of origin. Regulatory network analysis of transcription factors underscored the importance of BHLHE40 in luminal BC and luminal mature cells and KLF5 in basal-like tumors and luminal progenitor cells. Furthermore, we identify key genes defining the basal-like (SOX6 and KCNQ3) and luminal A/B (FAM155A and LRP1B) lineages. Exhausted CTLA4-expressing CD8+ T cells were enriched in basal-like BC, suggesting an altered means of immune dysfunction. These findings demonstrate analysis of paired transcription and chromatin accessibility at the single-cell level is a powerful tool for investigating cancer lineage and highlight transcriptional networks that define basal and luminal BC lineages.
Collapse
Affiliation(s)
- Michael D Iglesia
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
| | - Reyka G Jayasinghe
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
| | - Siqi Chen
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
| | - Nadezhda V Terekhanova
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
| | - John M Herndon
- Department of Surgery, Washington University in St. Louis, St. Louis, MO, USA
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Erik Storrs
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
| | - Alla Karpova
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
| | - Daniel Cui Zhou
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
| | - Nataly Naser Al Deen
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
| | - Andrew T Shinkle
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
| | - Rita Jui-Hsien Lu
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
| | - Wagma Caravan
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
| | - Andrew Houston
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
| | - Yanyan Zhao
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
| | - Kazuhito Sato
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
| | - Preet Lal
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Cherease Street
- Department of Surgery, Washington University in St. Louis, St. Louis, MO, USA
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Fernanda Martins Rodrigues
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
| | - Austin N Southard-Smith
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
| | - André Luiz N Targino da Costa
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
| | - Houxiang Zhu
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
| | - Chia-Kuei Mo
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
| | - Lisa Crowson
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
| | - Robert S Fulton
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
| | - Matthew A Wyczalkowski
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
| | - Catrina C Fronick
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
| | - Lucinda A Fulton
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
| | - Hua Sun
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
| | - Sherri R Davies
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | | | - Sara E Chasnoff
- Department of Surgery, Washington University in St. Louis, St. Louis, MO, USA
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Madelyn Carmody
- Department of Surgery, Washington University in St. Louis, St. Louis, MO, USA
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Candace Brooks
- Department of Surgery, Washington University in St. Louis, St. Louis, MO, USA
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Ruiyang Liu
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
| | - Michael C Wendl
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
- Department of Mathematics, Washington University in St. Louis, St. Louis, MO, USA
- Department of Genetics, Washington University in St. Louis, St. Louis, MO, USA
| | - Clara Oh
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
| | - Diane Bender
- Bursky Center for Human Immunology & Immunotherapy, Washington University in St. Louis, St. Louis, MO, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, USA
| | - Oscar Harari
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, USA
| | - Andrea Bredemeyer
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Kory Lavine
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO, USA
- Department of Developmental Biology, Washington University in St. Louis, St. Louis, MO, USA
| | - Ron Bose
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Julie Margenthaler
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Jason M Held
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Samuel Achilefu
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- Department of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Foluso Ademuyiwa
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Rebecca Aft
- Department of Surgery, Washington University in St. Louis, St. Louis, MO, USA
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
- John Cochran Veterans Hospital, St. Louis, MO, USA
| | - Cynthia Ma
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Graham A Colditz
- Department of Surgery, Washington University in St. Louis, St. Louis, MO, USA
- Division of Public Health Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Tao Ju
- Department of Computer Science and Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Stephen T Oh
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO, USA
| | - James Fitzpatrick
- Washington University Center for Cellular Imaging, Washington University in St. Louis, St. Louis, MO, USA
- Departments of Neuroscience and Cell Biology & Physiology, Washington University in St. Louis, St. Louis, MO, USA
| | - E Shelley Hwang
- Department of Surgery, Duke University Medical Center, Durham, NC, England
| | - Kooresh I Shoghi
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
- Department of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Milan G Chheda
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Deborah J Veis
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO, USA
| | - Feng Chen
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Ryan C Fields
- Department of Surgery, Washington University in St. Louis, St. Louis, MO, USA
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - William E Gillanders
- Department of Surgery, Washington University in St. Louis, St. Louis, MO, USA.
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA.
| | - Li Ding
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA.
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA.
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA.
- Department of Genetics, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
16
|
Murray CE, Kornepati AVR, Ontiveros C, Liao Y, de la Peña Avalos B, Rogers CM, Liu Z, Deng Y, Bai H, Kari S, Padron AS, Boyd JT, Reyes R, Clark CA, Svatek RS, Li R, Hu Y, Wang M, Conejo-Garcia JR, Byers LA, Ramkumar K, Sood AK, Lee JM, Burd CE, Vadlamudi RK, Gupta HB, Zhao W, Dray E, Sung P, Curiel TJ. Tumour-intrinsic PDL1 signals regulate the Chk2 DNA damage response in cancer cells and mediate resistance to Chk1 inhibitors. Mol Cancer 2024; 23:242. [PMID: 39478560 PMCID: PMC11523829 DOI: 10.1186/s12943-024-02147-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 10/05/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Aside from the canonical role of PDL1 as a tumour surface-expressed immune checkpoint molecule, tumour-intrinsic PDL1 signals regulate non-canonical immunopathological pathways mediating treatment resistance whose significance, mechanisms, and therapeutic targeting remain incompletely understood. Recent reports implicate tumour-intrinsic PDL1 signals in the DNA damage response (DDR), including promoting homologous recombination DNA damage repair and mRNA stability of DDR proteins, but many mechanistic details remain undefined. METHODS We genetically depleted PDL1 from transplantable mouse and human cancer cell lines to understand consequences of tumour-intrinsic PDL1 signals in the DNA damage response. We complemented this work with studies of primary human tumours and inducible mouse tumours. We developed novel approaches to show tumour-intrinsic PDL1 signals in specific subcellular locations. We pharmacologically depleted tumour PDL1 in vivo in mouse models with repurposed FDA-approved drugs for proof-of-concept clinical translation studies. RESULTS We show that tumour-intrinsic PDL1 promotes the checkpoint kinase-2 (Chk2)-mediated DNA damage response. Intracellular but not surface-expressed PDL1 controlled Chk2 protein content post-translationally and independently of PD1 by antagonising PIRH2 E3 ligase-mediated Chk2 polyubiquitination and protein degradation. Genetic tumour PDL1 depletion specifically reduced tumour Chk2 content but not ATM, ATR, or Chk1 DDR proteins, enhanced Chk1 inhibitor (Chk1i) synthetic lethality in vitro in diverse human and murine tumour models, and improved Chk1i efficacy in vivo. Pharmacologic tumour PDL1 depletion with cefepime or ceftazidime replicated genetic tumour PDL1 depletion by reducing tumour Chk2, inducing Chk1i synthetic lethality in a tumour PDL1-dependent manner, and reducing in vivo tumour growth when combined with Chk1i. CONCLUSIONS Our data challenge the prevailing surface PDL1 paradigm, elucidate important and previously unappreciated roles for tumour-intrinsic PDL1 in regulating the ATM/Chk2 DNA damage response axis and E3 ligase-mediated protein degradation, suggest tumour PDL1 as a biomarker for Chk1i efficacy, and support the rapid clinical potential of pharmacologic tumour PDL1 depletion to treat selected cancers.
Collapse
Affiliation(s)
- Clare E Murray
- Graduate School of Biomedical Sciences and Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Anand V R Kornepati
- Graduate School of Biomedical Sciences and Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
- Present address: Department of Internal Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Carlos Ontiveros
- Graduate School of Biomedical Sciences and Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Yiji Liao
- Dartmouth Cancer Center and Dartmouth Health, Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Bárbara de la Peña Avalos
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Cody M Rogers
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Zexuan Liu
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Yilun Deng
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Haiyan Bai
- Dartmouth Cancer Center and Dartmouth Health, Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Suresh Kari
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Alvaro S Padron
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Jacob T Boyd
- Graduate School of Biomedical Sciences and Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Ryan Reyes
- Graduate School of Biomedical Sciences and Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Curtis A Clark
- Graduate School of Biomedical Sciences and Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
- Present address: Department of Radiation Oncology, School of Medicine, University of Alabama Birmingham, Birmingham, USA
| | - Robert S Svatek
- Department of Urology, University of Texas Health San Antonio, San Antonio, TX, USA
- UT Health Mays Cancer Center, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Rong Li
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
- Present address: Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, DC, USA
| | - Yanfen Hu
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
- Present address: Department of Anatomy & Cell Biology, School of Medicine & Health Sciences, The George Washington University, Washington, DC, USA
| | - Meiling Wang
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX, USA
| | | | - Lauren A Byers
- Department of Thoracic/Head & Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kavya Ramkumar
- Department of Thoracic/Head & Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anil K Sood
- Department of Gynecologic Oncology & Reproductive Medicine, UT MD Anderson Cancer Center, Houston, TX, USA
| | - Jung-Min Lee
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Christin E Burd
- Departments of Molecular Genetics, Cancer Biology and Genetics, The Ohio State University, Columbus, OH, USA
| | - Ratna K Vadlamudi
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
- UT Health Mays Cancer Center, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Harshita B Gupta
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Weixing Zhao
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Eloïse Dray
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Patrick Sung
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX, USA
- UT Health Mays Cancer Center, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Tyler J Curiel
- Graduate School of Biomedical Sciences and Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA.
- Dartmouth Cancer Center and Dartmouth Health, Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA.
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA.
- UT Southwestern, Dallas, TX, USA.
- Dartmouth Health and Dartmouth Cancer Center, Lebanon, NH, USA.
- The Geisel School of Medicine at Dartmouth, Hanover, NH, USA.
- Department of Immunology, Dartmouth College, Hanover, NH, USA.
- Department of Integrative Immunobiology, Duke University, Durham, NC, USA.
| |
Collapse
|
17
|
Shang X, Zhang C, Lv Y, Zhang X, Guo K, Li H, Wang H. Patients with Extensive-Stage Small Cell Lung Cancer Harboring Less Than 4 Metastatic Sites May Benefit from Immune Checkpoint Inhibitor Rechallenge by Reshaping Tumor Microenvironment. Immunotargets Ther 2024; 13:571-583. [PMID: 39478941 PMCID: PMC11523948 DOI: 10.2147/itt.s483093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 10/22/2024] [Indexed: 11/02/2024] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) has prolonged survival in patients with extensive-stage small cell lung cancer (ES-SCLC) as first-line treatment. However, whether ICI rechallenge could bring survival benefit to patients with ES-SCLC following its failure as first-line treatment remains unknown. Therefore, we aim to address the issue and identify the cohort of patients that may derive such benefit. Methods Patients with ES-SCLC from both the IMpower133 study and Shandong Cancer Hospital and Institute (shanzhong cohort) who failed first-line ICI were included. Kaplan Meier analysis was performed to compare overall survival (OS). Both univariate and multivariate Cox regression analyses were conducted to identify factors affecting survival. Tumor immune cell infiltration was evaluated by the CIBERSORT algorithm and detected by multiplex immunofluorescence (mIF). Results A total of 125 ES-SCLC patients undergoing atezolizumab and 161 patients undergoing ICI as first-line treatment were recruited from IMpower133 and shanzhong cohort. Those receiving ICI rechallenge had a longer OS than those without in IMpower133 (P = 0.08) and shanzhong cohort (P = 0.013). In IMpower133 cohort, subgroup analyses found that patients with <4 metastatic sites derived more survival benefit from atezolizumab (P = 0.008). For patients with ES-SCLC harboring <4 metastatic sites, there was significant OS difference between atezolizumab versus non-atezolizumab as retreatment (P = 0.036). Moreover, for ES-SCLC patients with <4 metastatic sites, atezolizumab improved survival compared with non-atezolizumab (hazard ratio [HR]: 0.457; 95% CI: 0.256-0.817; P = 0.008). These findings were confirmed in shanzhong cohort. Those harboring <4 metastatic sites had fewer M2 macrophage and more CD4 naïve T cells infiltration, which was further confirmed by mIF of ES-SCLC samples from shanzhong cohort. Conclusion Our study provides rationale for ICI rechallenge among ES-SCLC patients with <4 metastatic sites, suggesting beneficial outcome by reshaping TME.
Collapse
Affiliation(s)
- Xiaoling Shang
- Shandong Cancer Hospital and Institute, Shandong University, Jinan, 250117, People’s Republic of China
| | - Chenyue Zhang
- Department of Integrated Therapy, Fudan University Shanghai Cancer Center, Shanghai Medical College, Shanghai, People’s Republic of China
| | - Yuanyuan Lv
- Department of Clinical Drug Research, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, People’s Republic of China
| | - Xiaoxiao Zhang
- Department of Clinical Drug Research, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, People’s Republic of China
| | - Kaiyue Guo
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Shandong University, Jinan, 250117, People’s Republic of China
| | - Huijuan Li
- Department of Clinical Drug Research, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, People’s Republic of China
| | - Haiyong Wang
- Department of Internal Medicine-Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, People’s Republic of China
| |
Collapse
|
18
|
Gu J, Xu J, Jiao A, Cai N, Gu T, Wu P, Cheng X, Chen B, Chen Y, Liu X. Comprehensive analysis of single-cell transcriptomics and genetic factors reveals the mechanisms and preventive strategies for the progression from pulmonary fibrosis to lung cancer. Int Immunopharmacol 2024; 140:112803. [PMID: 39094357 DOI: 10.1016/j.intimp.2024.112803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/24/2024] [Accepted: 07/23/2024] [Indexed: 08/04/2024]
Abstract
BACKGROUND Pulmonary fibrosis (PF) leads to excessive deposition of fibrous connective tissue in the lungs, increasing the risk of lung cancer due to the enhanced activity of fibroblasts (FBs). Fibroblast-mediated collagen fiber deposition creates a tumor-like microenvironment, laying the foundation for tumorigenesis. Clinically, numerous cases of lung cancer induced by pulmonary fibrosis have been observed. In recent years, the study of nucleotide point mutations, which provide more detailed insights than gene expression, has made significant advancements, offering new perspectives for clinical research. METHODS We initially employed Mendelian randomization to ascertain that the initial stage of lung cancer induced by PF belongs to small cell lung cancer (SCLC). Subsequently, pulmonary neuroendocrine cells (PNECs) were identified by using pseudo-time series analysis as cell clusters with carcinogenic potential. We categorized FBs into four groups according to their cellular metabolism, and then analyzed the cellular communication between FBs and PNECs, as well as changes in intracellular pathways of PNECs. Additionally, we examined the characteristic genome of FBs which is significantly associated with PF and investigated the impact of FBs on immune cells in the PF microenvironment. Finally, we explored strategies for preventing the progression from PF to lung cancer. RESULTS The genetic features of cells with carcinogenic potential in PF tissues were revealed, characterized by upregulation of Achaete-Scute Family BHLH Transcription Factor 1 (ASCL1), Homeobox B2 (HOXB2), Teashirt Zinc Finger Homeobox 2 (TSHZ2), Insulinoma-associated 1 (INSM1), and reduced activity of RE1 Silencing Transcription Factor (REST). FBs characterized by high glycolysis and low tricarboxylic acid (TCA) cycling played a key role in the progression of PF. The microenvironment of PF resembles the tumor microenvironment, providing a conducive immunosuppressive environment for the occurrence of cancer cells. In dendritic cells, rs9265808 is a susceptibility locus for progression from pulmonary fibrosis to lung cancer, mutations at this locus increase the expression of Complement Factor B (CFB), and excessive activation of the complement pathway is a crucial factor leading to lung cancer development in patients with pulmonary fibrosis. Ensuring adequate nutritional supply and physical function is one of the effective measures to prevent progression from pulmonary fibrosis to lung cancer. CONCLUSION CFB promotes lung cancer occurrence by inducing the accumulation and polarization of a large number of monocytes/macrophages in the lungs, driving disease progression by reducing the physical fitness of patients with pulmonary fibrosis.
Collapse
Affiliation(s)
- Jinghua Gu
- School of Life Sciences, Anhui Medical University, Hefei 230032, China; The First Clinical Medical College of Anhui Medical University, Hefei 230032, China; Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Henan Provincial Key Laboratory of Long Non-coding RNA and Cancer Metabolism, Translational Research Institute of Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jiansheng Xu
- The First Clinical Medical College of Anhui Medical University, Hefei 230032, China
| | - Annan Jiao
- The First Clinical Medical College of Anhui Medical University, Hefei 230032, China; The First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Ningning Cai
- The First Clinical Medical College of Anhui Medical University, Hefei 230032, China
| | - Tianrui Gu
- School of Pharmacy, Zhejiang University, Hangzhou 310058, China
| | - Pengcheng Wu
- School of Life Sciences, Anhui Medical University, Hefei 230032, China
| | - Xinyu Cheng
- School of Life Sciences, Anhui Medical University, Hefei 230032, China
| | - Bo Chen
- The First Clinical Medical College of Anhui Medical University, Hefei 230032, China; The First Affiliated Hospital of Anhui Medical University, Hefei 230032, China.
| | - Yang Chen
- The First Clinical Medical College of Anhui Medical University, Hefei 230032, China; The First Affiliated Hospital of Anhui Medical University, Hefei 230032, China.
| | - Xiaoying Liu
- School of Life Sciences, Anhui Medical University, Hefei 230032, China; Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Henan Provincial Key Laboratory of Long Non-coding RNA and Cancer Metabolism, Translational Research Institute of Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
19
|
Chen HJ, Gardner EE, Shah Y, Zhang K, Thakur A, Zhang C, Elemento O, Varmus H. FORMATION OF MALIGNANT, METASTATIC SMALL CELL LUNG CANCERS THROUGH OVERPRODUCTION OF cMYC PROTEIN IN TP53 AND RB1 DEPLETED PULMONARY NEUROENDOCRINE CELLS DERIVED FROM HUMAN EMBRYONIC STEM CELLS. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.06.561244. [PMID: 37873210 PMCID: PMC10592623 DOI: 10.1101/2023.10.06.561244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
We recently described our initial efforts to develop a model for small cell lung cancer (SCLC) derived from human embryonic stem cells (hESCs) that were differentiated to form pulmonary neuroendocrine cells (PNECs), a putative cell of origin for neuroendocrine-positive SCLC. Although reduced expression of the tumor suppressor genes TP53 and RB1 allowed the induced PNECs to form subcutaneous growths in immune-deficient mice, the tumors did not display the aggressive characteristics of SCLC seen in human patients. Here we report that the additional, doxycycline-regulated expression of a transgene encoding wild-type or mutant cMYC protein promotes rapid growth, invasion, and metastasis of these hESC-derived cells after injection into the renal capsule. Similar to others, we find that the addition of cMYC encourages the formation of the SCLC-N subtype, marked by high levels of NEUROD1 RNA. Using paired primary and metastatic samples for RNA sequencing, we observe that the subtype of SCLC does not change upon metastatic spread and that production of NEUROD1 is maintained. We also describe histological features of these malignant, SCLC-like tumors derived from hESCs and discuss potential uses of this model in efforts to control and better understand this recalcitrant neoplasm.
Collapse
Affiliation(s)
- Huanhuan Joyce Chen
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY
- The Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL
- The Ben May Department for Cancer Research, The University of Chicago, Chicago, IL
| | | | - Yajas Shah
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY
| | - Kui Zhang
- The Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL
- The Ben May Department for Cancer Research, The University of Chicago, Chicago, IL
| | - Abhimanyu Thakur
- The Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL
- The Ben May Department for Cancer Research, The University of Chicago, Chicago, IL
| | - Chen Zhang
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
| | - Olivier Elemento
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY
| | - Harold Varmus
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY
| |
Collapse
|
20
|
Huang X, Tian B, Ren Z, Zhang J, Yan W, Mo Y, Yuan J, Ma Y, Wang R, Liu R, Chen M, Yu J, Chen D. CD34 as a potential prognostic indicator for camrelizumab response in advanced non-small-cell lung cancer: insights from digital spatial profiling. Ther Adv Med Oncol 2024; 16:17588359241289671. [PMID: 39429466 PMCID: PMC11489950 DOI: 10.1177/17588359241289671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 09/19/2024] [Indexed: 10/22/2024] Open
Abstract
Background Given that only a small subset of patients with advanced non-small-cell lung cancer (aNSCLC) benefit from immune checkpoint inhibitors (ICIs), the effectiveness of ICIs is often compromised by the complex interplay within the tumor microenvironment (TME). Objectives To identify predictive biomarkers associated with ICI resistance at a multi-omics spatial level. Design A total of eight aNSCLC patients who received first-line anti-programmed cell death protein-1 (PD-1) monoclonal antibody camrelizumab at Shandong Cancer Hospital and Institute between 2021 and 2022 were included in the discovery cohort. An additional validation cohort of 45 samples from camrelizumab-treated aNSCLC patients was also enrolled. Methods NanoString GeoMx® digital spatial profiling was conducted at the transcriptomic and proteomic level within pan-cytokeratin (panCK+), CD45+, and CD68+ compartments. For validation, multiplex immunofluorescence (mIF) staining was performed. Results Distinct spatial expression patterns and levels of immune infiltration were observed between tumor and leukocyte compartments. Higher CD34 expression in the macrophage compartment correlated with poorer prognosis and response to camrelizumab (p < 0.05). mIF validation confirmed the association of elevated CD34 expression level with reduced progression-free survival (PFS; hazard ratio (HR) = 5.011, 95% confidence interval: 1.057-23.752, p = 0.042), outperforming traditional tumor markers in predictive accuracy. Conclusion Our findings identify CD34 as a novel spatial biomarker for anti-PD-1 therapy efficacy, potentially guiding the selection of aNSCLC patients who are more likely to benefit from ICI treatment. Trial registration ChiCTR2000040416.
Collapse
Affiliation(s)
- Xinyi Huang
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Baoqing Tian
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Ziyuan Ren
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Cheeloo College of Medicine, Shandong University Cancer Center, Jinan, Shandong, China
| | - Jingxin Zhang
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Cheeloo College of Medicine, Shandong University Cancer Center, Jinan, Shandong, China
| | - Weiwei Yan
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Cheeloo College of Medicine, Shandong University Cancer Center, Jinan, Shandong, China
| | - You Mo
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Jupeng Yuan
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yujiao Ma
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Cheeloo College of Medicine, Shandong University Cancer Center, Jinan, Shandong, China
| | - Ruiyang Wang
- Department of Oncology, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Rufei Liu
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Minxin Chen
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jinming Yu
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250000, Shandong, China
| | - Dawei Chen
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250000, Shandong, China
| |
Collapse
|
21
|
Zhao Q, Wang C, Huang W, Song Z, Lang Y, Zhu X. Potential immunologic and prognostic roles of CHRNA6 in SCLC and pan-cancer. Heliyon 2024; 10:e38572. [PMID: 39398083 PMCID: PMC11470509 DOI: 10.1016/j.heliyon.2024.e38572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/22/2024] [Accepted: 09/26/2024] [Indexed: 10/15/2024] Open
Abstract
Background Small cell lung cancer (SCLC) is considered the most malignant subtype of lung cancer, and it has a restricted range of therapeutic choices. The emergence of immunotherapy has offered new possibilities for patients with SCLC. However, the scarcity of clinical specimens has hampered the progress of clinical studies and we still face a shortage of dependable indicators to forecast the effectiveness of immunotherapy for SCLC. Methods In our study, we assessed the ImmuneScore and StromalScore of 81 SCLC samples obtained from the cBioPortal database. By comparing gene expression differences between the high and low immune scores groups, we identified 24 differentially expressed genes. Subsequently, an intersection was performed with genes that exhibited differential expression between normal and SCLC tissues, leading us to isolate the gene CHRNA6. To gain a deeper insight into the possible significance of CHRNA6 in SCLC, we singled out 50 genes that showed the most pronounced positive and negative associations with its expression. We then pinpointed hub genes for subsequent functional enrichment analyses by establishing a protein-protein interactions network. We additionally assessed the link between CHRNA6 expression in SCLC and characteristics of the immune microenvironment, along with the efficacy of immunotherapy, using the CIBERSORT, immunophenoscores (IPS), and tumor immune dysfunction and exclusion (TIDE) algorithms. Furthermore, we confirmed the prognostic impact of CHRNA6 expression in SCLC patients undergoing immunotherapy within a clinical cohort. Lastly, we obtained data from The Cancer Genome Atlas (TCGA) to investigate CHRNA6 expression in various tumors and its associations with genetic alterations, DNA methylation, copy number variation, clinicopathological characteristics, biological processes, immune microenvironment, prognosis, and drug sensitivity. Results In SCLC, we found that CHRNA6 function was associated with immune activation pathways such as antigen presentation processing and positive regulation of adaptive immune response, and that CHRNA6 demonstrated a strong correlation with immune cells infiltration. In addition, analysis of the clinical cohort revealed that patients with SCLC who exhibited elevated expression of CHRNA6 experienced better responses to immunotherapy. Our pan-cancer analysis disclosed that the expression of CHRNA6 is dysregulated in a multitude of cancers, potentially due to genetic mutations, copy number gains, and DNA demethylation. The gene set enrichment analysis (GSEA) outcomes indicated that CHRNA6 participates in immune responses and may play a positive immune regulatory role in most cancers. Furthermore, CHRNA6 has been observed to have a notable relationship with immune checkpoints, immunomodulators, immune cell infiltration, patient outcomes, and drug sensitivity across various cancers. Conclusions Our findings indicate that the CHRNA6 may act as a predictive indicator for SCLC patients receiving immunotherapy. The study also uncovers the aberrant expression of CHRNA6 in a range of human cancers and its potential roles in immunology and prognosis, offering novel perspectives for tailored cancer therapies.
Collapse
Affiliation(s)
- Qingqing Zhao
- Department of Respiratory and Critical Care Medicine, Southeast University, Zhongda Hospital, Nanjing, 210009, China
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Cong Wang
- Department of Pathology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Wucui Huang
- Department of Respiratory and Critical Care Medicine, Southeast University, Zhongda Hospital, Nanjing, 210009, China
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Zhongquan Song
- Department of Respiratory and Critical Care Medicine, Southeast University, Zhongda Hospital, Nanjing, 210009, China
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Yang Lang
- Department of Respiratory and Critical Care Medicine, Southeast University, Zhongda Hospital, Nanjing, 210009, China
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Xiaoli Zhu
- Department of Respiratory and Critical Care Medicine, Southeast University, Zhongda Hospital, Nanjing, 210009, China
- School of Medicine, Southeast University, Nanjing, 210009, China
| |
Collapse
|
22
|
Oh S, Koh J, Kim TM, Kim S, Youk J, Kim M, Keam B, Jeon YK, Ku JL, Kim DW, Chung DH, Heo DS. Transcriptomic Heterogeneity of EGFR-Mutant Non-Small Cell Lung Cancer Evolution Toward Small-Cell Lung Cancer. Clin Cancer Res 2024; 30:4729-4742. [PMID: 39150541 DOI: 10.1158/1078-0432.ccr-24-0160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 06/25/2024] [Accepted: 08/14/2024] [Indexed: 08/17/2024]
Abstract
PURPOSE Histologic transformation from EGFR-mutant non-small cell lung cancer (NSCLC) to small-cell lung cancer (SCLC) is a key mechanism of resistance to EGFR tyrosine kinase inhibitors (TKI). However, transcriptomic changes between NSCLC and transformed SCLC (t-SCLC) remain unexplored. EXPERIMENTAL DESIGN We conducted whole-transcriptome analysis of 59 regions of interest through the spatial profiling of formalin-fixed, paraffin-embedded tissues obtained from 10 patients (lung adenocarcinoma, 22; combined SCLC/NSCLC, 7; and t-SCLC, 30 regions of interests). Transcriptomic profiles and differentially expressed genes were compared between pre- and post-transformed tumors. RESULTS Following EGFR-TKI treatment, 93.7% (15/16) of t-SCLC components evolved into neuroendocrine-high subtypes (SCLC-A or SCLC-N). The transition to t-SCLC occurred regardless of EGFR-TKI treatment and EGFR mutational status, with a notable decrease in EGFR expression (P < 0.001) at both mRNA and protein levels. Pathway analysis revealed that gene overexpression was related to epigenetic alterations in t-SCLC. Interestingly, histone deacetylase inhibitors restored EGFR expression in SNU-2962A cells and their organoid model. The synergistic effects of third-generation EGFR-TKI osimertinib and the histone deacetylase inhibitor fimepinostat were validated in both in vitro and in vivo models. CONCLUSIONS Our study demonstrated that most t-SCLC cases showed neuronal subtypes with low EGFR expression. Differentially expressed gene analysis and t-SCLC preclinical models identified an epigenetic modifier as a promising treatment strategy for t-SCLC.
Collapse
Affiliation(s)
- Songji Oh
- Cancer Research Institute, Seoul National University, Seoul, South Korea
- Integrated Major in Innovative Medical Science, Seoul National University College of Medicine, Seoul, South Korea
| | - Jaemoon Koh
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Tae Min Kim
- Cancer Research Institute, Seoul National University, Seoul, South Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Soyeon Kim
- Cancer Research Institute, Seoul National University, Seoul, South Korea
- Integrated Major in Innovative Medical Science, Seoul National University College of Medicine, Seoul, South Korea
| | - Jeonghwan Youk
- Cancer Research Institute, Seoul National University, Seoul, South Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Miso Kim
- Cancer Research Institute, Seoul National University, Seoul, South Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Bhumsuk Keam
- Cancer Research Institute, Seoul National University, Seoul, South Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Yoon Kyung Jeon
- Cancer Research Institute, Seoul National University, Seoul, South Korea
- Integrated Major in Innovative Medical Science, Seoul National University College of Medicine, Seoul, South Korea
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Ja-Lok Ku
- Cancer Research Institute, Seoul National University, Seoul, South Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
| | - Dong-Wan Kim
- Cancer Research Institute, Seoul National University, Seoul, South Korea
- Integrated Major in Innovative Medical Science, Seoul National University College of Medicine, Seoul, South Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Doo Hyun Chung
- Cancer Research Institute, Seoul National University, Seoul, South Korea
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Dae Seog Heo
- Cancer Research Institute, Seoul National University, Seoul, South Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
23
|
Stewart CA, Diao L, Xi Y, Wang R, Ramkumar K, Serrano AG, Tanimoto A, Rodriguez BL, Morris BB, Shen L, Zhang B, Yang Y, Hamad SH, Cardnell RJ, Duarte A, Sahu M, Novegil VY, Weissman BE, Frumovitz M, Kalhor N, Soto LS, da Rocha P, Vokes N, Gibbons DL, Wang J, Heymach JV, Glisson B, Byers LA, Gay CM. YAP1 Status Defines Two Intrinsic Subtypes of LCNEC with Distinct Molecular Features and Therapeutic Vulnerabilities. Clin Cancer Res 2024; 30:4743-4754. [PMID: 39150543 PMCID: PMC11479841 DOI: 10.1158/1078-0432.ccr-24-0361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 05/03/2024] [Accepted: 08/14/2024] [Indexed: 08/17/2024]
Abstract
PURPOSE Large cell neuroendocrine carcinoma (LCNEC) is a high-grade neuroendocrine malignancy that, like small cell lung cancer (SCLC), is associated with the absence of druggable oncogenic drivers and dismal prognosis. In contrast to SCLC, however, there is little evidence to guide optimal treatment strategies, which are often adapted from SCLC and non-small cell lung cancer approaches. EXPERIMENTAL DESIGN To better define the biology of LCNEC, we analyzed cell line and patient genomic data and performed IHC and single-cell RNA sequencing of core needle biopsies from patients with LCNEC and preclinical models. RESULTS In this study, we demonstrate that the presence or absence of YAP1 distinguishes two subsets of LCNEC. The YAP1-high subset is mesenchymal and inflamed and is characterized, alongside TP53 mutations, by co-occurring alterations in CDKN2A/B and SMARCA4. Therapeutically, the YAP1-high subset demonstrates vulnerability to MEK- and AXL-targeting strategies, including a novel preclinical AXL chimeric antigen receptor-expressing T cell. Meanwhile, the YAP1-low subset is epithelial and immune-cold and more commonly features TP53 and RB1 co-mutations, similar to those observed in pure SCLC. Notably, the YAP1-low subset is also characterized by the expression of SCLC subtype-defining transcription factors, especially ASCL1 and NEUROD1, and as expected, given its transcriptional similarities to SCLC, exhibits putative vulnerabilities reminiscent of SCLC, including delta-like ligand 3 and CD56 targeting, as is with novel preclinical delta-like ligand 3 and CD56 chimeric antigen receptor-expressing T cells, and DNA damage repair inhibition. CONCLUSIONS YAP1 defines distinct subsets of LCNEC with unique biology. These findings highlight the potential for YAP1 to guide personalized treatment strategies for LCNEC.
Collapse
MESH Headings
- Humans
- YAP-Signaling Proteins/genetics
- YAP-Signaling Proteins/metabolism
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Lung Neoplasms/genetics
- Lung Neoplasms/pathology
- Lung Neoplasms/metabolism
- Carcinoma, Neuroendocrine/genetics
- Carcinoma, Neuroendocrine/pathology
- Carcinoma, Neuroendocrine/metabolism
- Cell Line, Tumor
- Carcinoma, Large Cell/genetics
- Carcinoma, Large Cell/pathology
- Carcinoma, Large Cell/metabolism
- Carcinoma, Large Cell/therapy
- Animals
- Mutation
- Biomarkers, Tumor/genetics
- Mice
- Gene Expression Regulation, Neoplastic
- Prognosis
Collapse
Affiliation(s)
- C. Allison Stewart
- Department of Thoracic/Head & Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lixia Diao
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yuanxin Xi
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Runsheng Wang
- Department of Thoracic/Head & Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kavya Ramkumar
- Department of Thoracic/Head & Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alejandra G. Serrano
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Azusa Tanimoto
- Department of Thoracic/Head & Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - B. Leticia Rodriguez
- Department of Thoracic/Head & Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Benjamin B. Morris
- Department of Thoracic/Head & Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Li Shen
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bingnan Zhang
- Department of Thoracic/Head & Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yan Yang
- Department of Thoracic/Head & Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Samera H. Hamad
- Department of Surgery, Cooper Medical School of Rowan University, Camden, NJ USA
| | - Robert J. Cardnell
- Department of Thoracic/Head & Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alberto Duarte
- Department of Thoracic/Head & Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Moushumi Sahu
- Department of Thoracic/Head & Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Veronica Y. Novegil
- Department of Thoracic/Head & Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bernard E. Weissman
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Michael Frumovitz
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Neda Kalhor
- Department of Thoracic/Head & Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Luisa Solis Soto
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Pedro da Rocha
- Department of Medical Oncology, Vall d’Hebron University Hospital, Barcelona, Spain
| | - Natalie Vokes
- Department of Thoracic/Head & Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Don L. Gibbons
- Department of Thoracic/Head & Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jing Wang
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John V. Heymach
- Department of Thoracic/Head & Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bonnie Glisson
- Department of Thoracic/Head & Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lauren Averett Byers
- Department of Thoracic/Head & Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Carl M. Gay
- Department of Thoracic/Head & Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
24
|
Meyer ML, Peters S, Mok TS, Lam S, Yang PC, Aggarwal C, Brahmer J, Dziadziuszko R, Felip E, Ferris A, Forde PM, Gray J, Gros L, Halmos B, Herbst R, Jänne PA, Johnson BE, Kelly K, Leighl NB, Liu S, Lowy I, Marron TU, Paz-Ares L, Rizvi N, Rudin CM, Shum E, Stahel R, Trunova N, Ujhazy P, Bunn PA, Hirsch FR. Lung Cancer Research and Treatment: Global Perspectives and Strategic Calls to Action. Ann Oncol 2024:S0923-7534(24)04055-9. [PMID: 39413875 DOI: 10.1016/j.annonc.2024.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 10/02/2024] [Accepted: 10/04/2024] [Indexed: 10/18/2024] Open
Abstract
BACKGROUND Lung cancer remains a critical public health issue, presenting multifaceted challenges in prevention, diagnosis, and treatment. This article aims to review the current landscape of lung cancer research and management, delineate the persistent challenges, and outline pragmatic solutions. MATERIALS AND METHODS Global experts from academia, regulatory agencies such as the Food and Drug Administration (FDA) and the European Medicines Agency (EMA), the National Cancer Institute (NCI), professional societies, the pharmaceutical and biotech industries, and patient advocacy groups were gathered by the New York Lung Cancer Foundation to review the state of the art in lung cancer and to formulate calls to action. RESULTS Improving lung cancer management and research involves promoting tobacco cessation, identifying individuals at risk who could benefit from early detection programs, and addressing treatment-related toxicities. Efforts should focus on conducting well-designed trials to determine the optimal treatment sequence. Research into innovative biomarkers and therapies is crucial for more personalized treatment. Ensuring access to appropriate care for all patients, whether enrolled in clinical trials or not, must remain a priority. CONCLUSIONS Lung cancer is a major health burden worldwide, and its treatment has become increasingly complex over the past two decades. Improvement in lung cancer management and research requires unified messaging and global collaboration, expanded education, and greater access to screening, biomarker testing, treatment, as well as increased representativeness, participation, and diversity in clinical trials.
Collapse
Affiliation(s)
- M-L Meyer
- Icahn School of Medicine and Center for Thoracic Oncology, Tisch Cancer Institute at Mount Sinai, New York, USA
| | - S Peters
- Department of Oncology, University Hospital (CHUV), Lausanne, Switzerland
| | - T S Mok
- State Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong, China
| | - S Lam
- Department of Integrative Oncology, BC Cancer and the University of British Columbia, Vancouver, Canada
| | - P-C Yang
- Department of Internal Medicine, National Taiwan University College of Medicine, Taiwan
| | - C Aggarwal
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - J Brahmer
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Kimmel Cancer Center, Baltimore, USA
| | - R Dziadziuszko
- Medical University of Gdansk, Department of Oncology and Radiotherapy, Gdansk, Poland
| | - E Felip
- Medical Oncology Department, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - A Ferris
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins Kimmel Cancer Center, Baltimore, USA
| | | | - J Gray
- Department of Radiology, Mount Sinai Hospital, New York, USA
| | - L Gros
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, USA
| | - B Halmos
- Department of Oncology, MD Montefiore Einstein Comprehensive Cancer Center, New York, USA
| | - R Herbst
- Department of Medical Oncology, Yale Comprehensive Cancer Center, New Haven, USA
| | - P A Jänne
- Lowe Center for Thoracic Oncology, Department of Medical Oncology, Dana Farber Cancer Institute, Boston, USA
| | - B E Johnson
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - K Kelly
- International Association for the Study of Lung Cancer, Denver, CO, USA
| | - N B Leighl
- Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - S Liu
- Division of Medicine, Georgetown University, Washington, USA
| | - I Lowy
- Regeneron Pharmaceuticals, Inc., Tarrytown, USA
| | - T U Marron
- Early Phase Trials Unit and Center for Thoracic Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - L Paz-Ares
- Department of Oncology; Hospital Universitario 12 de Octubre, Madrid, Spain
| | - N Rizvi
- Synthekine, Inc. Menlo Park, USA
| | - C M Rudin
- Departments of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | - E Shum
- Division of Medical Oncology, Department of Medicine, Perlmutter Cancer Center, New York University Grossman School of Medicine, New York, USA
| | - R Stahel
- ETOP IBCSG Partners Foundation, Bern, Switzerland
| | - N Trunova
- Global Medical Affairs, Genmab, Princeton, USA
| | - P Ujhazy
- National Cancer Institute, Rockville, USA
| | - P A Bunn
- Division of Medical Oncology, University of Colorado School of Medicine, Aurora, USA
| | - F R Hirsch
- Icahn School of Medicine and Thoracic Oncology Center, Tisch Cancer Institute at Mount Sinai, New York, USA.
| |
Collapse
|
25
|
Zhai W, Yu Y, Wu H, Zhang Q, Chen Y, Yang Y, Fan Y. Real-world efficacy of PD-1/PD-L1 inhibitors in patients with advanced pulmonary neuroendocrine carcinoma: a single-center analysis. Ther Adv Med Oncol 2024; 16:17588359241288130. [PMID: 39410959 PMCID: PMC11475206 DOI: 10.1177/17588359241288130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 09/13/2024] [Indexed: 10/19/2024] Open
Abstract
Background Immunotherapy blocking programmed death-1 (PD-1)/programmed death ligand-1 (PD-L1) has revolutionized the treatment of extensive-stage small-cell lung cancer (SCLC), but only with limited real-world efficacy data; evidence from immunotherapy for other pulmonary neuroendocrine carcinoma (PNEC) is scarce. Objective The purpose of this study is to evaluate the efficacy of receiving PD-1/PD-L1 inhibitors in patients with advanced PNEC and explore factors related to survival prognosis, providing clues for treatment for patients with advanced PNEC. Methods In all, 203 patients with advanced PNEC who received PD-1/PD-L1 inhibitors between January 2019 and December 2021 were retrospectively analyzed. Kaplan-Meier curves were constructed for progression-free survival (PFS) and overall survival (OS). Results For the 203 patients, the objective response rate (ORR) was 48.3%, the disease control rate (DCR) was 83.3%, the median PFS (mPFS) was 6.0 months, and the median OS (mOS) was 13.1 months. Among them, the histology was 166 SCLC, 13 large-cell neuroendocrine carcinoma, and 24 other unspecified PNEC. Histologically, no significant difference was observed in PFS (p = 0.240) or OS (p = 0.845). In first-line (1L) treatment (N = 125), patients received chemoimmunotherapy and had an ORR of 64.8%, DCR of 92.0%, mPFS of 6.6 months, and mOS of 14.9 months. In second-line (2L) or later-line setting, the ORR, DCR, mPFS, and mOS were 21.8%, 69.2%, 4.4, and 9.4 months; immunotherapy plus small-molecule antiangiogenic agents showed significantly greater PFS than immunotherapy monotherapy or chemoimmunotherapy (6.4 vs 1.4 vs 3.7 months, p = 0.041). Patients without liver metastasis had superior PFS (7.0 vs 5.1 months, p < 0.001) and OS (19.2 vs 9.6 months, p < 0.001) than those with liver metastasis. Conclusion In clinical practice, PD-1/PD-L1 inhibitors are effective in patients with advanced PNEC, regardless of the pathological histology. The efficacy of 1L immunochemotherapy is worthy of recognition, and the addition of small-molecule antiangiogenic agents to immunotherapy in 2L or later-line treatment provides a better survival trend. Design Retrospective study.
Collapse
Affiliation(s)
- Wanchen Zhai
- Department of Oncology, The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Ying Yu
- Department of Oncology, The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Haicheng Wu
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Postgraduate Training Base Alliance of Wenzhou Medical University, Wenzhou, China
| | - Qian Zhang
- Department of Oncology, The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Yunfei Chen
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Postgraduate Training Base Alliance of Wenzhou Medical University, Wenzhou, China
| | - Yehao Yang
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Postgraduate Training Base Alliance of Wenzhou Medical University, Wenzhou, China
| | - Yun Fan
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, No. 1 East Banshan Road, Gongshu District, Hangzhou, Zhejiang 310022, China
| |
Collapse
|
26
|
Saalfeld FC, Möller J, Christopoulos P, Wenzel C, Rasokat A, Wang XA, Vathiotis I, König D, Illini O, Grohé C, Wiesweg M, Wesseler C, Schubart C, Pelusi N, Rohde G, Overbeck TR, Kirfel J, Alt J, Kauffmann-Guerrero D, Griesinger F, Kulhavy J, Allgäuer M, Klimova A, Schütz M, Aust DE, Hochmair MJ, Rothschild SI, Syrigos KN, Veluswamy R, Michels S, Stenzinger A, Jöhrens K, Wermke M. Small cell transformation in EGFR-mutated non-small cell lung cancer: DLL3 expression and efficacy of immune checkpoint inhibitors or tyrosine kinase inhibitors combined with chemotherapy. Eur J Cancer 2024; 213:115065. [PMID: 39423775 DOI: 10.1016/j.ejca.2024.115065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/29/2024] [Accepted: 10/05/2024] [Indexed: 10/21/2024]
Abstract
INTRODUCTION Small cell transformation (SCT) is a typical mechanism of adaptive resistance to third generation epidermal growth factor receptor inhibitors (EGFRi) which have become the standard of care for EGFR-driven non-small cell lung cancer (EGFR+ NSCLC). Little is known about the optimal management of SCT patients. This study aimed to compare outcomes under platinum/etoposide chemotherapy alone (chemo) or in combination with EGFR inhibitors (EGFRi+chemo) or immune checkpoint inhibitors (ICI+chemo). In addition, DLL3 expression was explored as potential novel therapeutic target. METHODS We conducted a retrospective study on patients with EGFR+ NSCLC and SCT treated at 19 centers in Europe and the United States. A total of 47 patients were included of whom 17 received chemo, 20 ICI+chemo, and 10 EGFRi+chemo. We analyzed DLL3 expression by immunohistochemistry. RESULTS In the entire cohort, median overall survival (OS) from start of first SCT therapy was 11 months (95 % confidence interval [95 %CI] 9.1-12.9) and median progression-free survival (PFS) was 5 months (95 %CI 4.2-5.8). Median PFS was similar in all three groups (chemo and ICI+chemo 4 months, EGFRi+chemo 6 months), and 12-months PFS was 12 % (95 %CI 2 %-31 %), 13 % (95 %CI 0 %-43 %), and 0 % for ICI+chemo, EGFRi+chemo, and chemo, respectively. Median OS in the ICI+chemo group was 13 months (95 %CI 5.5-20.5) compared to 10 months (95 %CI 7.6-12.4) with chemo and EGFRi+chemo (95 %CI 8.1-11.9), respectively. Before and after SCT, 0 % and 93 % of tumors were DLL3-positive. CONCLUSIONS Our results suggest that ICI+chemo and DLL3-targeting agents are worth further exploration in EGFR+ NSCLC undergoing SCT. PRESENTED ELSEWHERE Part of this work has been presented at ESMO annual meeting in Madrid, Spain in October 2023 (Poster 1336 P).
Collapse
Affiliation(s)
- Felix Carl Saalfeld
- Clinic for Internal Medicine I, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany; National Center for Tumor Diseases, Dresden, Germany; National Network Genomic Medicine Lung Cancer (nNGM), Germany
| | - Johanna Möller
- Clinic for Internal Medicine I, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany; National Center for Tumor Diseases, Dresden, Germany
| | - Petros Christopoulos
- Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany; Translational Lung Research Center Heidelberg (TLRC-H), member of the German Center for Lung Research (DZL), Heidelberg, Germany; National Network Genomic Medicine Lung Cancer (nNGM), Germany
| | - Carina Wenzel
- National Center for Tumor Diseases, Dresden, Germany; Department for Pathology, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany; National Network Genomic Medicine Lung Cancer (nNGM), Germany
| | - Anna Rasokat
- Lung Cancer Group Cologne, Department I for Internal Medicine and Center for Integrated Oncology Aachen Bonn Cologne Dusseldorf, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany; National Network Genomic Medicine Lung Cancer (nNGM), Germany
| | - Xuejun Alice Wang
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Ioannis Vathiotis
- Third Department of Internal Medicine, Sotiria Hospital, National, and Kapodistrian University of Athens, Athens, Greece
| | - David König
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
| | - Oliver Illini
- Department of Respiratory and Critical Care Medicine, Klinik Floridsdorf, Vienna Healthcare Group, Vienna, Austria; Karl Landsteiner Institute of Lung Research and Pulmonary Oncology, Vienna, Austria
| | - Christian Grohé
- Department of Respiratory Diseases - ELK, Lindenberger Weg 27, 13125 Berlin, Germany; National Network Genomic Medicine Lung Cancer (nNGM), Germany
| | - Marcel Wiesweg
- West German Cancer Center, Department of Medical Oncology, University Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany; National Network Genomic Medicine Lung Cancer (nNGM), Germany
| | - Claas Wesseler
- Department of Pneumology, Asklepios Tumorzentrum Hamburg, Klinikum Harburg, Hamburg, Germany
| | - Christoph Schubart
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; National Network Genomic Medicine Lung Cancer (nNGM), Germany
| | - Natalie Pelusi
- Institute of Pathology, University Hospital, University of Bonn, Venusberg-Campus 1, Gebäude 62, 53127 Bonn, Germany; National Network Genomic Medicine Lung Cancer (nNGM), Germany
| | - Gernot Rohde
- Goethe University Frankfurt, University Hospital, Medical Clinic I, Department of Respiratory Medicine, Frankfurt/Main, Germany
| | - Tobias R Overbeck
- Department of Hematology and Medical Oncology, University Medical Center Göttingen, Göttingen University, Göttingen, Germany; National Network Genomic Medicine Lung Cancer (nNGM), Germany
| | - Jutta Kirfel
- Institute of Pathology, UKSH Campus Lübeck, University Hospital Schleswig-Holstein, Ratzenburger Allee 160, Haus V50, 23538 Lübeck, Germany; National Network Genomic Medicine Lung Cancer (nNGM), Germany
| | - Jürgen Alt
- Department of Hematology and Medical Oncology, University Medical Center Mainz, Mainz, Germany; National Network Genomic Medicine Lung Cancer (nNGM), Germany
| | - Diego Kauffmann-Guerrero
- Division of Respiratory Medicine and Thoracic Oncology, Department of Medicine V, Thoracic Oncology Center Munich, University Hospital, University of Munich (LMU), Germany; National Network Genomic Medicine Lung Cancer (nNGM), Germany
| | - Frank Griesinger
- Department of Hematology and Oncology, Pius Hospital, University Medicine Oldenburg, Oldenburg, Germany; National Network Genomic Medicine Lung Cancer (nNGM), Germany
| | - Jonas Kulhavy
- Translational Oncology/Early Clinical Trial Unit (ECTU), Bavarian Cancer Research Center, National Center for Tumor Diseases, Comprehensive Cancer Center Mainfranken and University Hospital Würzburg, Würzburg, Germany; National Network Genomic Medicine Lung Cancer (nNGM), Germany
| | - Michael Allgäuer
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany; National Network Genomic Medicine Lung Cancer (nNGM), Germany
| | - Anna Klimova
- Core Unit for Data Management and Analytics, National Center for Tumor Diseases, Dresden, Germany
| | - Maret Schütz
- National Center for Tumor Diseases, Dresden, Germany; Department for Pathology, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany; National Network Genomic Medicine Lung Cancer (nNGM), Germany
| | - Daniela E Aust
- National Center for Tumor Diseases, Dresden, Germany; Department for Pathology, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany; National Network Genomic Medicine Lung Cancer (nNGM), Germany
| | - Maximilian J Hochmair
- Department of Respiratory and Critical Care Medicine, Klinik Floridsdorf, Vienna Healthcare Group, Vienna, Austria; Karl Landsteiner Institute of Lung Research and Pulmonary Oncology, Vienna, Austria
| | - Sacha I Rothschild
- Cantonal Hospital Baden, Department Internal Medicine, Center of Oncology & Hematology, Baden, Switzerland
| | - Konstantinos N Syrigos
- Third Department of Internal Medicine, Sotiria Hospital, National, and Kapodistrian University of Athens, Athens, Greece
| | - Rajwanth Veluswamy
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Sebastian Michels
- Lung Cancer Group Cologne, Department I for Internal Medicine and Center for Integrated Oncology Aachen Bonn Cologne Dusseldorf, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany; National Network Genomic Medicine Lung Cancer (nNGM), Germany
| | - Albrecht Stenzinger
- Translational Lung Research Center Heidelberg (TLRC-H), member of the German Center for Lung Research (DZL), Heidelberg, Germany; Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany; National Network Genomic Medicine Lung Cancer (nNGM), Germany
| | - Korinna Jöhrens
- National Center for Tumor Diseases, Dresden, Germany; Department for Pathology, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany; Department for Pathology, Klinikum Chemnitz gGmbH, Chemnitz, Germany
| | - Martin Wermke
- Clinic for Internal Medicine I, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany; National Center for Tumor Diseases, Dresden, Germany; National Network Genomic Medicine Lung Cancer (nNGM), Germany.
| |
Collapse
|
27
|
Yu T, Lok BH. Strategies to Target Chemoradiotherapy Resistance in Small Cell Lung Cancer. Cancers (Basel) 2024; 16:3438. [PMID: 39456533 PMCID: PMC11506711 DOI: 10.3390/cancers16203438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/04/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Small cell lung cancer (SCLC) is a lethal form of lung cancer with few treatment options and a high rate of relapse. While SCLC is initially sensitive to first-line DNA-damaging chemo- and radiotherapy, relapse disease is almost universally therapy-resistant. As a result, there has been interest in understanding the mechanisms of therapeutic resistance in this disease. Conclusions: Progress has been made in elucidating these mechanisms, particularly as they relate to the DNA damage response and SCLC differentiation and transformation, leading to many clinical trials investigating new therapies and combinations. Yet there remain many gaps in our understanding, such as the effect of epigenetics or the tumor microenvironment on treatment response, and no single mechanism has been found to be ubiquitous, suggesting a significant heterogeneity in the mechanisms of acquired resistance. Nevertheless, the advancement of techniques in the laboratory and the clinic will improve our ability to study this disease, especially in patient populations, and identify methods to surmount therapeutic resistance.
Collapse
Affiliation(s)
- Tony Yu
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, 101 College Street, Toronto, ON M5G 1L7, Canada
| | - Benjamin H. Lok
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, 101 College Street, Toronto, ON M5G 1L7, Canada
- Radiation Medicine Program, Princess Margaret Cancer Centre, 610 University Ave, Toronto, ON M5G 2M9, Canada
- Department of Radiation Oncology, Temerty Faculty of Medicine, University of Toronto, 149 College Street, Toronto, ON M5T 1P5, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, 6 Queen’s Park Crescent, Toronto, ON M5S 3H2, Canada
| |
Collapse
|
28
|
Umemura S, Udagawa H, Ikeda T, Murakami H, Daga H, Toyozawa R, Kozuki T, Sakakibara-Konishi J, Ohe Y, Morise M, Kato T, Shingyoji M, Hara S, Furuya N, Teranishi S, Takata S, Miyamoto S, Nakachi I, Wakabayashi M, Nomura S, Sato A, Ishii G, Tsuchihara K, Sugiyama E, Kirita K, Sakai T, Shibata Y, Izumi H, Nosaki K, Zenke Y, Matsumoto S, Yoh K, Niho S, Goto K. Clinical Significance of a Prospective Large Genomic Screening for SCLC: The Genetic Classification and a Biomarker-Driven Phase 2 Trial of Gedatolisib. J Thorac Oncol 2024:S1556-0864(24)02378-5. [PMID: 39395663 DOI: 10.1016/j.jtho.2024.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 09/17/2024] [Accepted: 10/05/2024] [Indexed: 10/14/2024]
Abstract
INTRODUCTION SCLC has been treated as a single entity resulting in limited survival improvement. Developing effective tools for guiding appropriate therapeutic strategies is crucial. METHODS A total of 1035 SCLCs were prospectively analyzed by a genomic screening platform: LC-SCRUM-Asia. Fresh frozen tumor samples were subjected to a next-generation sequencing system enabling the integrative analysis of cancer-related genes. A phase 2 trial of gedatolisib for SCLC with PI3K/AKT/mTOR pathway mutations was conducted based on this screening. RESULTS On the basis of the treatment outcomes and therapeutic targets, the following five distinct genetic subgroups were identified in SCLC: NSCLC-subgroup (genetic alterations associated with NSCLC, 8.5%); Hotspot-subgroup (targetable hotspot mutations common in tumors, 3.0%); PI3K-subgroup (PI3K/AKT/mTOR pathway mutations, 7.4%); MYC-subgroup (MYC family amplifications, 13.0%); and HME-subgroup (mutations in the histone-modifying enzymes, 17.6%). The NSCLC-subgroup (hazard ratio = 1.57; 95% confidence interval: 1.22-2.03) and MYC-subgroup (hazard ratio = 1.56; 95% confidence interval: 1.26-1.93) had significantly shorter progression-free survivals after first-line platinum-based treatment. The Hotspot-subgroup and MYC-subgroup were candidates for novel targeted therapies. The HME-subgroup had a favorable survival in patients who received programmed cell death (ligand) 1 inhibitor-based therapies (p = 0.005, log-rank test) regardless of some overlap with other subgroups. There were 15 patients enrolled into the phase 2 trial of gedatolisib in the PI3K-subgroup, and the overall response rate and the disease control rate were 6.7% and 20%, respectively. The MYC-subgroup or NSCLC-subgroup was associated with unfavorable clinical outcomes in this trial. CONCLUSIONS Molecular classification of SCLC by genetic approach is beneficial for predicting the treatment outcomes and effectively guiding the clinical choices.
Collapse
Affiliation(s)
- Shigeki Umemura
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hibiki Udagawa
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takaya Ikeda
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Haruyasu Murakami
- Division of Thoracic Oncology, Shizuoka Cancer Center, Shizuoka, Japan
| | - Haruko Daga
- Department of Medical Oncology, Osaka City General Hospital, Osaka, Japan
| | - Ryo Toyozawa
- Department of Thoracic Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Toshiyuki Kozuki
- Department of Thoracic Oncology and Medicine, National Hospital Organization Shikoku Cancer Center, Matsuyama, Japan
| | - Jun Sakakibara-Konishi
- Department of Respiratory Medicine, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Yuichiro Ohe
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Masahiro Morise
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Terufumi Kato
- Department of Thoracic Oncology, Kanagawa Cancer Center, Yokohama, Japan
| | | | - Satoshi Hara
- Department of Respiratory Medicine, Itami City Hospital, Itami, Japan
| | - Naoki Furuya
- Division of Respiratory Medicine, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Shuhei Teranishi
- Respiratory Disease Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Saori Takata
- Department of Respiratory Medicine, Kyorin University School of Medicine, Mitaka, Japan
| | - Shingo Miyamoto
- Department of Medical Oncology, Japanese Red Cross Medical Center, Tokyo, Japan
| | - Ichiro Nakachi
- Department of Internal Medicine, Saiseikai Utsunomiya Hospital, Utsunomiya, Japan
| | - Masashi Wakabayashi
- Clinical Research Support Office, National Cancer Center Hospital East, Chiba, Japan
| | - Shogo Nomura
- Clinical Research Support Office, National Cancer Center Hospital East, Chiba, Japan; Department of Biostatistics and Bioinformatics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Akihiro Sato
- Clinical Research Support Office, National Cancer Center Hospital East, Chiba, Japan
| | - Genichiro Ishii
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Chiba, Japan
| | - Katsuya Tsuchihara
- Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Eri Sugiyama
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Keisuke Kirita
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Tetsuya Sakai
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yuji Shibata
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hiroki Izumi
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Kaname Nosaki
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yoshitaka Zenke
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Shingo Matsumoto
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Kiyotaka Yoh
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Seiji Niho
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Koichi Goto
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan.
| |
Collapse
|
29
|
Cheng Y, Spigel DR, Cho BC, Laktionov KK, Fang J, Chen Y, Zenke Y, Lee KH, Wang Q, Navarro A, Bernabe R, Buchmeier EL, Chang JWC, Shiraishi Y, Goksu SS, Badzio A, Shi A, Daniel DB, Hoa NTT, Zemanova M, Mann H, Gowda H, Jiang H, Senan S. Durvalumab after Chemoradiotherapy in Limited-Stage Small-Cell Lung Cancer. N Engl J Med 2024; 391:1313-1327. [PMID: 39268857 DOI: 10.1056/nejmoa2404873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
BACKGROUND Adjuvant therapy with durvalumab, with or without tremelimumab, may have efficacy in patients with limited-stage small-cell lung cancer who do not have disease progression after standard concurrent platinum-based chemoradiotherapy. METHODS In a phase 3, double-blind, randomized, placebo-controlled trial, we assigned patients to receive durvalumab at a dose of 1500 mg, durvalumab (1500 mg) plus tremelimumab at a dose of 75 mg (four doses only), or placebo every 4 weeks for up to 24 months. Randomization was stratified according to disease stage (I or II vs. III) and receipt of prophylactic cranial irradiation (yes vs. no). Results of the first planned interim analysis of the two primary end points of overall survival and progression-free survival (assessed on the basis of blinded independent central review according to the Response Evaluation Criteria in Solid Tumors, version 1.1) with durvalumab as compared with placebo (data cutoff date, January 15, 2024) are reported; results in the durvalumab-tremelimumab group remain blinded. RESULTS A total of 264 patients were assigned to the durvalumab group, 200 to the durvalumab-tremelimumab group, and 266 to the placebo group. Durvalumab therapy led to significantly longer overall survival than placebo (median, 55.9 months [95% confidence interval {CI}, 37.3 to not reached] vs. 33.4 months [95% CI, 25.5 to 39.9]; hazard ratio for death, 0.73; 98.321% CI, 0.54 to 0.98; P = 0.01), as well as to significantly longer progression-free survival (median 16.6 months [95% CI, 10.2 to 28.2] vs. 9.2 months [95% CI, 7.4 to 12.9]; hazard ratio for progression or death, 0.76; 97.195% CI, 0.59 to 0.98; P = 0.02). The incidence of adverse events with a maximum grade of 3 or 4 was 24.4% among patients receiving durvalumab and 24.2% among patients receiving placebo; adverse events led to discontinuation in 16.4% and 10.6% of the patients, respectively, and led to death in 2.7% and 1.9%. Pneumonitis or radiation pneumonitis with a maximum grade of 3 or 4 occurred in 3.1% of the patients in the durvalumab group and in 2.6% of those in the placebo group. CONCLUSIONS Adjuvant therapy with durvalumab led to significantly longer overall survival and progression-free survival than placebo among patients with limited-stage small-cell lung cancer. (Funded by AstraZeneca; ADRIATIC ClinicalTrials.gov number, NCT03703297.).
Collapse
Affiliation(s)
- Ying Cheng
- From Jilin Cancer Hospital, Changchun (Y. Cheng), Department II of Thoracic Oncology (J.F.) and the Department of Radiation Oncology (A.S.), Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, and the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou (Q.W.) - all in China; Sarah Cannon Research Institute (D.R.S.) and Tennessee Oncology (D.B.D.) - both in Nashville; Yonsei Cancer Center, Yonsei University College of Medicine, Seoul (B.C.C.), and Chungbuk National University Hospital, Cheongju (K.H.L.) - both in South Korea; Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology" of the Ministry of Health of the Russian Federation, Moscow (K.K.L.); Cancer and Hematology Centers of Western Michigan, Grand Rapids (Y. Chen); National Cancer Center Hospital East, Kashiwa (Y.Z.), and Kyushu University Hospital, Fukuoka (Y.S.) - both in Japan; Hospital Vall d'Hebron and Vall d'Hebron Institute of Oncology, Barcelona (A.N.), and Hospital Universitario Virgen del Rocio, Seville (R.B.) - both in Spain; Hospitals of the City of Cologne, Cologne, Germany (E.L.B.); Chang Gung Medical Foundation-Linkou Branch, Taoyuan, Taiwan (J.W.-C.C.); Akdeniz University Medical Faculty, Antalya, Turkey (S.S.G.); Medical University of Gdansk, Gdansk, Poland (A.B.); National Cancer Hospital, Hanoi, Vietnam (N.T.T.H.); First Faculty of Medicine, Charles University, and General University Hospital, Prague, Czech Republic (M.Z.); AstraZeneca, Cambridge, United Kingdom (H.M.); AstraZeneca, Gaithersburg, MD (H.G., H.J.); and the Department of Radiation Oncology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam (S.S.)
| | - David R Spigel
- From Jilin Cancer Hospital, Changchun (Y. Cheng), Department II of Thoracic Oncology (J.F.) and the Department of Radiation Oncology (A.S.), Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, and the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou (Q.W.) - all in China; Sarah Cannon Research Institute (D.R.S.) and Tennessee Oncology (D.B.D.) - both in Nashville; Yonsei Cancer Center, Yonsei University College of Medicine, Seoul (B.C.C.), and Chungbuk National University Hospital, Cheongju (K.H.L.) - both in South Korea; Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology" of the Ministry of Health of the Russian Federation, Moscow (K.K.L.); Cancer and Hematology Centers of Western Michigan, Grand Rapids (Y. Chen); National Cancer Center Hospital East, Kashiwa (Y.Z.), and Kyushu University Hospital, Fukuoka (Y.S.) - both in Japan; Hospital Vall d'Hebron and Vall d'Hebron Institute of Oncology, Barcelona (A.N.), and Hospital Universitario Virgen del Rocio, Seville (R.B.) - both in Spain; Hospitals of the City of Cologne, Cologne, Germany (E.L.B.); Chang Gung Medical Foundation-Linkou Branch, Taoyuan, Taiwan (J.W.-C.C.); Akdeniz University Medical Faculty, Antalya, Turkey (S.S.G.); Medical University of Gdansk, Gdansk, Poland (A.B.); National Cancer Hospital, Hanoi, Vietnam (N.T.T.H.); First Faculty of Medicine, Charles University, and General University Hospital, Prague, Czech Republic (M.Z.); AstraZeneca, Cambridge, United Kingdom (H.M.); AstraZeneca, Gaithersburg, MD (H.G., H.J.); and the Department of Radiation Oncology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam (S.S.)
| | - Byoung Chul Cho
- From Jilin Cancer Hospital, Changchun (Y. Cheng), Department II of Thoracic Oncology (J.F.) and the Department of Radiation Oncology (A.S.), Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, and the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou (Q.W.) - all in China; Sarah Cannon Research Institute (D.R.S.) and Tennessee Oncology (D.B.D.) - both in Nashville; Yonsei Cancer Center, Yonsei University College of Medicine, Seoul (B.C.C.), and Chungbuk National University Hospital, Cheongju (K.H.L.) - both in South Korea; Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology" of the Ministry of Health of the Russian Federation, Moscow (K.K.L.); Cancer and Hematology Centers of Western Michigan, Grand Rapids (Y. Chen); National Cancer Center Hospital East, Kashiwa (Y.Z.), and Kyushu University Hospital, Fukuoka (Y.S.) - both in Japan; Hospital Vall d'Hebron and Vall d'Hebron Institute of Oncology, Barcelona (A.N.), and Hospital Universitario Virgen del Rocio, Seville (R.B.) - both in Spain; Hospitals of the City of Cologne, Cologne, Germany (E.L.B.); Chang Gung Medical Foundation-Linkou Branch, Taoyuan, Taiwan (J.W.-C.C.); Akdeniz University Medical Faculty, Antalya, Turkey (S.S.G.); Medical University of Gdansk, Gdansk, Poland (A.B.); National Cancer Hospital, Hanoi, Vietnam (N.T.T.H.); First Faculty of Medicine, Charles University, and General University Hospital, Prague, Czech Republic (M.Z.); AstraZeneca, Cambridge, United Kingdom (H.M.); AstraZeneca, Gaithersburg, MD (H.G., H.J.); and the Department of Radiation Oncology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam (S.S.)
| | - Konstantin K Laktionov
- From Jilin Cancer Hospital, Changchun (Y. Cheng), Department II of Thoracic Oncology (J.F.) and the Department of Radiation Oncology (A.S.), Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, and the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou (Q.W.) - all in China; Sarah Cannon Research Institute (D.R.S.) and Tennessee Oncology (D.B.D.) - both in Nashville; Yonsei Cancer Center, Yonsei University College of Medicine, Seoul (B.C.C.), and Chungbuk National University Hospital, Cheongju (K.H.L.) - both in South Korea; Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology" of the Ministry of Health of the Russian Federation, Moscow (K.K.L.); Cancer and Hematology Centers of Western Michigan, Grand Rapids (Y. Chen); National Cancer Center Hospital East, Kashiwa (Y.Z.), and Kyushu University Hospital, Fukuoka (Y.S.) - both in Japan; Hospital Vall d'Hebron and Vall d'Hebron Institute of Oncology, Barcelona (A.N.), and Hospital Universitario Virgen del Rocio, Seville (R.B.) - both in Spain; Hospitals of the City of Cologne, Cologne, Germany (E.L.B.); Chang Gung Medical Foundation-Linkou Branch, Taoyuan, Taiwan (J.W.-C.C.); Akdeniz University Medical Faculty, Antalya, Turkey (S.S.G.); Medical University of Gdansk, Gdansk, Poland (A.B.); National Cancer Hospital, Hanoi, Vietnam (N.T.T.H.); First Faculty of Medicine, Charles University, and General University Hospital, Prague, Czech Republic (M.Z.); AstraZeneca, Cambridge, United Kingdom (H.M.); AstraZeneca, Gaithersburg, MD (H.G., H.J.); and the Department of Radiation Oncology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam (S.S.)
| | - Jian Fang
- From Jilin Cancer Hospital, Changchun (Y. Cheng), Department II of Thoracic Oncology (J.F.) and the Department of Radiation Oncology (A.S.), Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, and the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou (Q.W.) - all in China; Sarah Cannon Research Institute (D.R.S.) and Tennessee Oncology (D.B.D.) - both in Nashville; Yonsei Cancer Center, Yonsei University College of Medicine, Seoul (B.C.C.), and Chungbuk National University Hospital, Cheongju (K.H.L.) - both in South Korea; Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology" of the Ministry of Health of the Russian Federation, Moscow (K.K.L.); Cancer and Hematology Centers of Western Michigan, Grand Rapids (Y. Chen); National Cancer Center Hospital East, Kashiwa (Y.Z.), and Kyushu University Hospital, Fukuoka (Y.S.) - both in Japan; Hospital Vall d'Hebron and Vall d'Hebron Institute of Oncology, Barcelona (A.N.), and Hospital Universitario Virgen del Rocio, Seville (R.B.) - both in Spain; Hospitals of the City of Cologne, Cologne, Germany (E.L.B.); Chang Gung Medical Foundation-Linkou Branch, Taoyuan, Taiwan (J.W.-C.C.); Akdeniz University Medical Faculty, Antalya, Turkey (S.S.G.); Medical University of Gdansk, Gdansk, Poland (A.B.); National Cancer Hospital, Hanoi, Vietnam (N.T.T.H.); First Faculty of Medicine, Charles University, and General University Hospital, Prague, Czech Republic (M.Z.); AstraZeneca, Cambridge, United Kingdom (H.M.); AstraZeneca, Gaithersburg, MD (H.G., H.J.); and the Department of Radiation Oncology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam (S.S.)
| | - Yuanbin Chen
- From Jilin Cancer Hospital, Changchun (Y. Cheng), Department II of Thoracic Oncology (J.F.) and the Department of Radiation Oncology (A.S.), Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, and the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou (Q.W.) - all in China; Sarah Cannon Research Institute (D.R.S.) and Tennessee Oncology (D.B.D.) - both in Nashville; Yonsei Cancer Center, Yonsei University College of Medicine, Seoul (B.C.C.), and Chungbuk National University Hospital, Cheongju (K.H.L.) - both in South Korea; Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology" of the Ministry of Health of the Russian Federation, Moscow (K.K.L.); Cancer and Hematology Centers of Western Michigan, Grand Rapids (Y. Chen); National Cancer Center Hospital East, Kashiwa (Y.Z.), and Kyushu University Hospital, Fukuoka (Y.S.) - both in Japan; Hospital Vall d'Hebron and Vall d'Hebron Institute of Oncology, Barcelona (A.N.), and Hospital Universitario Virgen del Rocio, Seville (R.B.) - both in Spain; Hospitals of the City of Cologne, Cologne, Germany (E.L.B.); Chang Gung Medical Foundation-Linkou Branch, Taoyuan, Taiwan (J.W.-C.C.); Akdeniz University Medical Faculty, Antalya, Turkey (S.S.G.); Medical University of Gdansk, Gdansk, Poland (A.B.); National Cancer Hospital, Hanoi, Vietnam (N.T.T.H.); First Faculty of Medicine, Charles University, and General University Hospital, Prague, Czech Republic (M.Z.); AstraZeneca, Cambridge, United Kingdom (H.M.); AstraZeneca, Gaithersburg, MD (H.G., H.J.); and the Department of Radiation Oncology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam (S.S.)
| | - Yoshitaka Zenke
- From Jilin Cancer Hospital, Changchun (Y. Cheng), Department II of Thoracic Oncology (J.F.) and the Department of Radiation Oncology (A.S.), Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, and the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou (Q.W.) - all in China; Sarah Cannon Research Institute (D.R.S.) and Tennessee Oncology (D.B.D.) - both in Nashville; Yonsei Cancer Center, Yonsei University College of Medicine, Seoul (B.C.C.), and Chungbuk National University Hospital, Cheongju (K.H.L.) - both in South Korea; Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology" of the Ministry of Health of the Russian Federation, Moscow (K.K.L.); Cancer and Hematology Centers of Western Michigan, Grand Rapids (Y. Chen); National Cancer Center Hospital East, Kashiwa (Y.Z.), and Kyushu University Hospital, Fukuoka (Y.S.) - both in Japan; Hospital Vall d'Hebron and Vall d'Hebron Institute of Oncology, Barcelona (A.N.), and Hospital Universitario Virgen del Rocio, Seville (R.B.) - both in Spain; Hospitals of the City of Cologne, Cologne, Germany (E.L.B.); Chang Gung Medical Foundation-Linkou Branch, Taoyuan, Taiwan (J.W.-C.C.); Akdeniz University Medical Faculty, Antalya, Turkey (S.S.G.); Medical University of Gdansk, Gdansk, Poland (A.B.); National Cancer Hospital, Hanoi, Vietnam (N.T.T.H.); First Faculty of Medicine, Charles University, and General University Hospital, Prague, Czech Republic (M.Z.); AstraZeneca, Cambridge, United Kingdom (H.M.); AstraZeneca, Gaithersburg, MD (H.G., H.J.); and the Department of Radiation Oncology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam (S.S.)
| | - Ki Hyeong Lee
- From Jilin Cancer Hospital, Changchun (Y. Cheng), Department II of Thoracic Oncology (J.F.) and the Department of Radiation Oncology (A.S.), Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, and the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou (Q.W.) - all in China; Sarah Cannon Research Institute (D.R.S.) and Tennessee Oncology (D.B.D.) - both in Nashville; Yonsei Cancer Center, Yonsei University College of Medicine, Seoul (B.C.C.), and Chungbuk National University Hospital, Cheongju (K.H.L.) - both in South Korea; Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology" of the Ministry of Health of the Russian Federation, Moscow (K.K.L.); Cancer and Hematology Centers of Western Michigan, Grand Rapids (Y. Chen); National Cancer Center Hospital East, Kashiwa (Y.Z.), and Kyushu University Hospital, Fukuoka (Y.S.) - both in Japan; Hospital Vall d'Hebron and Vall d'Hebron Institute of Oncology, Barcelona (A.N.), and Hospital Universitario Virgen del Rocio, Seville (R.B.) - both in Spain; Hospitals of the City of Cologne, Cologne, Germany (E.L.B.); Chang Gung Medical Foundation-Linkou Branch, Taoyuan, Taiwan (J.W.-C.C.); Akdeniz University Medical Faculty, Antalya, Turkey (S.S.G.); Medical University of Gdansk, Gdansk, Poland (A.B.); National Cancer Hospital, Hanoi, Vietnam (N.T.T.H.); First Faculty of Medicine, Charles University, and General University Hospital, Prague, Czech Republic (M.Z.); AstraZeneca, Cambridge, United Kingdom (H.M.); AstraZeneca, Gaithersburg, MD (H.G., H.J.); and the Department of Radiation Oncology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam (S.S.)
| | - Qiming Wang
- From Jilin Cancer Hospital, Changchun (Y. Cheng), Department II of Thoracic Oncology (J.F.) and the Department of Radiation Oncology (A.S.), Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, and the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou (Q.W.) - all in China; Sarah Cannon Research Institute (D.R.S.) and Tennessee Oncology (D.B.D.) - both in Nashville; Yonsei Cancer Center, Yonsei University College of Medicine, Seoul (B.C.C.), and Chungbuk National University Hospital, Cheongju (K.H.L.) - both in South Korea; Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology" of the Ministry of Health of the Russian Federation, Moscow (K.K.L.); Cancer and Hematology Centers of Western Michigan, Grand Rapids (Y. Chen); National Cancer Center Hospital East, Kashiwa (Y.Z.), and Kyushu University Hospital, Fukuoka (Y.S.) - both in Japan; Hospital Vall d'Hebron and Vall d'Hebron Institute of Oncology, Barcelona (A.N.), and Hospital Universitario Virgen del Rocio, Seville (R.B.) - both in Spain; Hospitals of the City of Cologne, Cologne, Germany (E.L.B.); Chang Gung Medical Foundation-Linkou Branch, Taoyuan, Taiwan (J.W.-C.C.); Akdeniz University Medical Faculty, Antalya, Turkey (S.S.G.); Medical University of Gdansk, Gdansk, Poland (A.B.); National Cancer Hospital, Hanoi, Vietnam (N.T.T.H.); First Faculty of Medicine, Charles University, and General University Hospital, Prague, Czech Republic (M.Z.); AstraZeneca, Cambridge, United Kingdom (H.M.); AstraZeneca, Gaithersburg, MD (H.G., H.J.); and the Department of Radiation Oncology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam (S.S.)
| | - Alejandro Navarro
- From Jilin Cancer Hospital, Changchun (Y. Cheng), Department II of Thoracic Oncology (J.F.) and the Department of Radiation Oncology (A.S.), Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, and the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou (Q.W.) - all in China; Sarah Cannon Research Institute (D.R.S.) and Tennessee Oncology (D.B.D.) - both in Nashville; Yonsei Cancer Center, Yonsei University College of Medicine, Seoul (B.C.C.), and Chungbuk National University Hospital, Cheongju (K.H.L.) - both in South Korea; Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology" of the Ministry of Health of the Russian Federation, Moscow (K.K.L.); Cancer and Hematology Centers of Western Michigan, Grand Rapids (Y. Chen); National Cancer Center Hospital East, Kashiwa (Y.Z.), and Kyushu University Hospital, Fukuoka (Y.S.) - both in Japan; Hospital Vall d'Hebron and Vall d'Hebron Institute of Oncology, Barcelona (A.N.), and Hospital Universitario Virgen del Rocio, Seville (R.B.) - both in Spain; Hospitals of the City of Cologne, Cologne, Germany (E.L.B.); Chang Gung Medical Foundation-Linkou Branch, Taoyuan, Taiwan (J.W.-C.C.); Akdeniz University Medical Faculty, Antalya, Turkey (S.S.G.); Medical University of Gdansk, Gdansk, Poland (A.B.); National Cancer Hospital, Hanoi, Vietnam (N.T.T.H.); First Faculty of Medicine, Charles University, and General University Hospital, Prague, Czech Republic (M.Z.); AstraZeneca, Cambridge, United Kingdom (H.M.); AstraZeneca, Gaithersburg, MD (H.G., H.J.); and the Department of Radiation Oncology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam (S.S.)
| | - Reyes Bernabe
- From Jilin Cancer Hospital, Changchun (Y. Cheng), Department II of Thoracic Oncology (J.F.) and the Department of Radiation Oncology (A.S.), Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, and the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou (Q.W.) - all in China; Sarah Cannon Research Institute (D.R.S.) and Tennessee Oncology (D.B.D.) - both in Nashville; Yonsei Cancer Center, Yonsei University College of Medicine, Seoul (B.C.C.), and Chungbuk National University Hospital, Cheongju (K.H.L.) - both in South Korea; Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology" of the Ministry of Health of the Russian Federation, Moscow (K.K.L.); Cancer and Hematology Centers of Western Michigan, Grand Rapids (Y. Chen); National Cancer Center Hospital East, Kashiwa (Y.Z.), and Kyushu University Hospital, Fukuoka (Y.S.) - both in Japan; Hospital Vall d'Hebron and Vall d'Hebron Institute of Oncology, Barcelona (A.N.), and Hospital Universitario Virgen del Rocio, Seville (R.B.) - both in Spain; Hospitals of the City of Cologne, Cologne, Germany (E.L.B.); Chang Gung Medical Foundation-Linkou Branch, Taoyuan, Taiwan (J.W.-C.C.); Akdeniz University Medical Faculty, Antalya, Turkey (S.S.G.); Medical University of Gdansk, Gdansk, Poland (A.B.); National Cancer Hospital, Hanoi, Vietnam (N.T.T.H.); First Faculty of Medicine, Charles University, and General University Hospital, Prague, Czech Republic (M.Z.); AstraZeneca, Cambridge, United Kingdom (H.M.); AstraZeneca, Gaithersburg, MD (H.G., H.J.); and the Department of Radiation Oncology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam (S.S.)
| | - Eva Lotte Buchmeier
- From Jilin Cancer Hospital, Changchun (Y. Cheng), Department II of Thoracic Oncology (J.F.) and the Department of Radiation Oncology (A.S.), Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, and the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou (Q.W.) - all in China; Sarah Cannon Research Institute (D.R.S.) and Tennessee Oncology (D.B.D.) - both in Nashville; Yonsei Cancer Center, Yonsei University College of Medicine, Seoul (B.C.C.), and Chungbuk National University Hospital, Cheongju (K.H.L.) - both in South Korea; Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology" of the Ministry of Health of the Russian Federation, Moscow (K.K.L.); Cancer and Hematology Centers of Western Michigan, Grand Rapids (Y. Chen); National Cancer Center Hospital East, Kashiwa (Y.Z.), and Kyushu University Hospital, Fukuoka (Y.S.) - both in Japan; Hospital Vall d'Hebron and Vall d'Hebron Institute of Oncology, Barcelona (A.N.), and Hospital Universitario Virgen del Rocio, Seville (R.B.) - both in Spain; Hospitals of the City of Cologne, Cologne, Germany (E.L.B.); Chang Gung Medical Foundation-Linkou Branch, Taoyuan, Taiwan (J.W.-C.C.); Akdeniz University Medical Faculty, Antalya, Turkey (S.S.G.); Medical University of Gdansk, Gdansk, Poland (A.B.); National Cancer Hospital, Hanoi, Vietnam (N.T.T.H.); First Faculty of Medicine, Charles University, and General University Hospital, Prague, Czech Republic (M.Z.); AstraZeneca, Cambridge, United Kingdom (H.M.); AstraZeneca, Gaithersburg, MD (H.G., H.J.); and the Department of Radiation Oncology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam (S.S.)
| | - John Wen-Cheng Chang
- From Jilin Cancer Hospital, Changchun (Y. Cheng), Department II of Thoracic Oncology (J.F.) and the Department of Radiation Oncology (A.S.), Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, and the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou (Q.W.) - all in China; Sarah Cannon Research Institute (D.R.S.) and Tennessee Oncology (D.B.D.) - both in Nashville; Yonsei Cancer Center, Yonsei University College of Medicine, Seoul (B.C.C.), and Chungbuk National University Hospital, Cheongju (K.H.L.) - both in South Korea; Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology" of the Ministry of Health of the Russian Federation, Moscow (K.K.L.); Cancer and Hematology Centers of Western Michigan, Grand Rapids (Y. Chen); National Cancer Center Hospital East, Kashiwa (Y.Z.), and Kyushu University Hospital, Fukuoka (Y.S.) - both in Japan; Hospital Vall d'Hebron and Vall d'Hebron Institute of Oncology, Barcelona (A.N.), and Hospital Universitario Virgen del Rocio, Seville (R.B.) - both in Spain; Hospitals of the City of Cologne, Cologne, Germany (E.L.B.); Chang Gung Medical Foundation-Linkou Branch, Taoyuan, Taiwan (J.W.-C.C.); Akdeniz University Medical Faculty, Antalya, Turkey (S.S.G.); Medical University of Gdansk, Gdansk, Poland (A.B.); National Cancer Hospital, Hanoi, Vietnam (N.T.T.H.); First Faculty of Medicine, Charles University, and General University Hospital, Prague, Czech Republic (M.Z.); AstraZeneca, Cambridge, United Kingdom (H.M.); AstraZeneca, Gaithersburg, MD (H.G., H.J.); and the Department of Radiation Oncology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam (S.S.)
| | - Yoshimasa Shiraishi
- From Jilin Cancer Hospital, Changchun (Y. Cheng), Department II of Thoracic Oncology (J.F.) and the Department of Radiation Oncology (A.S.), Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, and the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou (Q.W.) - all in China; Sarah Cannon Research Institute (D.R.S.) and Tennessee Oncology (D.B.D.) - both in Nashville; Yonsei Cancer Center, Yonsei University College of Medicine, Seoul (B.C.C.), and Chungbuk National University Hospital, Cheongju (K.H.L.) - both in South Korea; Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology" of the Ministry of Health of the Russian Federation, Moscow (K.K.L.); Cancer and Hematology Centers of Western Michigan, Grand Rapids (Y. Chen); National Cancer Center Hospital East, Kashiwa (Y.Z.), and Kyushu University Hospital, Fukuoka (Y.S.) - both in Japan; Hospital Vall d'Hebron and Vall d'Hebron Institute of Oncology, Barcelona (A.N.), and Hospital Universitario Virgen del Rocio, Seville (R.B.) - both in Spain; Hospitals of the City of Cologne, Cologne, Germany (E.L.B.); Chang Gung Medical Foundation-Linkou Branch, Taoyuan, Taiwan (J.W.-C.C.); Akdeniz University Medical Faculty, Antalya, Turkey (S.S.G.); Medical University of Gdansk, Gdansk, Poland (A.B.); National Cancer Hospital, Hanoi, Vietnam (N.T.T.H.); First Faculty of Medicine, Charles University, and General University Hospital, Prague, Czech Republic (M.Z.); AstraZeneca, Cambridge, United Kingdom (H.M.); AstraZeneca, Gaithersburg, MD (H.G., H.J.); and the Department of Radiation Oncology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam (S.S.)
| | - Sema Sezgin Goksu
- From Jilin Cancer Hospital, Changchun (Y. Cheng), Department II of Thoracic Oncology (J.F.) and the Department of Radiation Oncology (A.S.), Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, and the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou (Q.W.) - all in China; Sarah Cannon Research Institute (D.R.S.) and Tennessee Oncology (D.B.D.) - both in Nashville; Yonsei Cancer Center, Yonsei University College of Medicine, Seoul (B.C.C.), and Chungbuk National University Hospital, Cheongju (K.H.L.) - both in South Korea; Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology" of the Ministry of Health of the Russian Federation, Moscow (K.K.L.); Cancer and Hematology Centers of Western Michigan, Grand Rapids (Y. Chen); National Cancer Center Hospital East, Kashiwa (Y.Z.), and Kyushu University Hospital, Fukuoka (Y.S.) - both in Japan; Hospital Vall d'Hebron and Vall d'Hebron Institute of Oncology, Barcelona (A.N.), and Hospital Universitario Virgen del Rocio, Seville (R.B.) - both in Spain; Hospitals of the City of Cologne, Cologne, Germany (E.L.B.); Chang Gung Medical Foundation-Linkou Branch, Taoyuan, Taiwan (J.W.-C.C.); Akdeniz University Medical Faculty, Antalya, Turkey (S.S.G.); Medical University of Gdansk, Gdansk, Poland (A.B.); National Cancer Hospital, Hanoi, Vietnam (N.T.T.H.); First Faculty of Medicine, Charles University, and General University Hospital, Prague, Czech Republic (M.Z.); AstraZeneca, Cambridge, United Kingdom (H.M.); AstraZeneca, Gaithersburg, MD (H.G., H.J.); and the Department of Radiation Oncology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam (S.S.)
| | - Andrzej Badzio
- From Jilin Cancer Hospital, Changchun (Y. Cheng), Department II of Thoracic Oncology (J.F.) and the Department of Radiation Oncology (A.S.), Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, and the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou (Q.W.) - all in China; Sarah Cannon Research Institute (D.R.S.) and Tennessee Oncology (D.B.D.) - both in Nashville; Yonsei Cancer Center, Yonsei University College of Medicine, Seoul (B.C.C.), and Chungbuk National University Hospital, Cheongju (K.H.L.) - both in South Korea; Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology" of the Ministry of Health of the Russian Federation, Moscow (K.K.L.); Cancer and Hematology Centers of Western Michigan, Grand Rapids (Y. Chen); National Cancer Center Hospital East, Kashiwa (Y.Z.), and Kyushu University Hospital, Fukuoka (Y.S.) - both in Japan; Hospital Vall d'Hebron and Vall d'Hebron Institute of Oncology, Barcelona (A.N.), and Hospital Universitario Virgen del Rocio, Seville (R.B.) - both in Spain; Hospitals of the City of Cologne, Cologne, Germany (E.L.B.); Chang Gung Medical Foundation-Linkou Branch, Taoyuan, Taiwan (J.W.-C.C.); Akdeniz University Medical Faculty, Antalya, Turkey (S.S.G.); Medical University of Gdansk, Gdansk, Poland (A.B.); National Cancer Hospital, Hanoi, Vietnam (N.T.T.H.); First Faculty of Medicine, Charles University, and General University Hospital, Prague, Czech Republic (M.Z.); AstraZeneca, Cambridge, United Kingdom (H.M.); AstraZeneca, Gaithersburg, MD (H.G., H.J.); and the Department of Radiation Oncology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam (S.S.)
| | - Anhui Shi
- From Jilin Cancer Hospital, Changchun (Y. Cheng), Department II of Thoracic Oncology (J.F.) and the Department of Radiation Oncology (A.S.), Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, and the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou (Q.W.) - all in China; Sarah Cannon Research Institute (D.R.S.) and Tennessee Oncology (D.B.D.) - both in Nashville; Yonsei Cancer Center, Yonsei University College of Medicine, Seoul (B.C.C.), and Chungbuk National University Hospital, Cheongju (K.H.L.) - both in South Korea; Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology" of the Ministry of Health of the Russian Federation, Moscow (K.K.L.); Cancer and Hematology Centers of Western Michigan, Grand Rapids (Y. Chen); National Cancer Center Hospital East, Kashiwa (Y.Z.), and Kyushu University Hospital, Fukuoka (Y.S.) - both in Japan; Hospital Vall d'Hebron and Vall d'Hebron Institute of Oncology, Barcelona (A.N.), and Hospital Universitario Virgen del Rocio, Seville (R.B.) - both in Spain; Hospitals of the City of Cologne, Cologne, Germany (E.L.B.); Chang Gung Medical Foundation-Linkou Branch, Taoyuan, Taiwan (J.W.-C.C.); Akdeniz University Medical Faculty, Antalya, Turkey (S.S.G.); Medical University of Gdansk, Gdansk, Poland (A.B.); National Cancer Hospital, Hanoi, Vietnam (N.T.T.H.); First Faculty of Medicine, Charles University, and General University Hospital, Prague, Czech Republic (M.Z.); AstraZeneca, Cambridge, United Kingdom (H.M.); AstraZeneca, Gaithersburg, MD (H.G., H.J.); and the Department of Radiation Oncology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam (S.S.)
| | - Davey B Daniel
- From Jilin Cancer Hospital, Changchun (Y. Cheng), Department II of Thoracic Oncology (J.F.) and the Department of Radiation Oncology (A.S.), Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, and the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou (Q.W.) - all in China; Sarah Cannon Research Institute (D.R.S.) and Tennessee Oncology (D.B.D.) - both in Nashville; Yonsei Cancer Center, Yonsei University College of Medicine, Seoul (B.C.C.), and Chungbuk National University Hospital, Cheongju (K.H.L.) - both in South Korea; Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology" of the Ministry of Health of the Russian Federation, Moscow (K.K.L.); Cancer and Hematology Centers of Western Michigan, Grand Rapids (Y. Chen); National Cancer Center Hospital East, Kashiwa (Y.Z.), and Kyushu University Hospital, Fukuoka (Y.S.) - both in Japan; Hospital Vall d'Hebron and Vall d'Hebron Institute of Oncology, Barcelona (A.N.), and Hospital Universitario Virgen del Rocio, Seville (R.B.) - both in Spain; Hospitals of the City of Cologne, Cologne, Germany (E.L.B.); Chang Gung Medical Foundation-Linkou Branch, Taoyuan, Taiwan (J.W.-C.C.); Akdeniz University Medical Faculty, Antalya, Turkey (S.S.G.); Medical University of Gdansk, Gdansk, Poland (A.B.); National Cancer Hospital, Hanoi, Vietnam (N.T.T.H.); First Faculty of Medicine, Charles University, and General University Hospital, Prague, Czech Republic (M.Z.); AstraZeneca, Cambridge, United Kingdom (H.M.); AstraZeneca, Gaithersburg, MD (H.G., H.J.); and the Department of Radiation Oncology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam (S.S.)
| | - Nguyen Thi Thai Hoa
- From Jilin Cancer Hospital, Changchun (Y. Cheng), Department II of Thoracic Oncology (J.F.) and the Department of Radiation Oncology (A.S.), Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, and the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou (Q.W.) - all in China; Sarah Cannon Research Institute (D.R.S.) and Tennessee Oncology (D.B.D.) - both in Nashville; Yonsei Cancer Center, Yonsei University College of Medicine, Seoul (B.C.C.), and Chungbuk National University Hospital, Cheongju (K.H.L.) - both in South Korea; Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology" of the Ministry of Health of the Russian Federation, Moscow (K.K.L.); Cancer and Hematology Centers of Western Michigan, Grand Rapids (Y. Chen); National Cancer Center Hospital East, Kashiwa (Y.Z.), and Kyushu University Hospital, Fukuoka (Y.S.) - both in Japan; Hospital Vall d'Hebron and Vall d'Hebron Institute of Oncology, Barcelona (A.N.), and Hospital Universitario Virgen del Rocio, Seville (R.B.) - both in Spain; Hospitals of the City of Cologne, Cologne, Germany (E.L.B.); Chang Gung Medical Foundation-Linkou Branch, Taoyuan, Taiwan (J.W.-C.C.); Akdeniz University Medical Faculty, Antalya, Turkey (S.S.G.); Medical University of Gdansk, Gdansk, Poland (A.B.); National Cancer Hospital, Hanoi, Vietnam (N.T.T.H.); First Faculty of Medicine, Charles University, and General University Hospital, Prague, Czech Republic (M.Z.); AstraZeneca, Cambridge, United Kingdom (H.M.); AstraZeneca, Gaithersburg, MD (H.G., H.J.); and the Department of Radiation Oncology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam (S.S.)
| | - Milada Zemanova
- From Jilin Cancer Hospital, Changchun (Y. Cheng), Department II of Thoracic Oncology (J.F.) and the Department of Radiation Oncology (A.S.), Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, and the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou (Q.W.) - all in China; Sarah Cannon Research Institute (D.R.S.) and Tennessee Oncology (D.B.D.) - both in Nashville; Yonsei Cancer Center, Yonsei University College of Medicine, Seoul (B.C.C.), and Chungbuk National University Hospital, Cheongju (K.H.L.) - both in South Korea; Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology" of the Ministry of Health of the Russian Federation, Moscow (K.K.L.); Cancer and Hematology Centers of Western Michigan, Grand Rapids (Y. Chen); National Cancer Center Hospital East, Kashiwa (Y.Z.), and Kyushu University Hospital, Fukuoka (Y.S.) - both in Japan; Hospital Vall d'Hebron and Vall d'Hebron Institute of Oncology, Barcelona (A.N.), and Hospital Universitario Virgen del Rocio, Seville (R.B.) - both in Spain; Hospitals of the City of Cologne, Cologne, Germany (E.L.B.); Chang Gung Medical Foundation-Linkou Branch, Taoyuan, Taiwan (J.W.-C.C.); Akdeniz University Medical Faculty, Antalya, Turkey (S.S.G.); Medical University of Gdansk, Gdansk, Poland (A.B.); National Cancer Hospital, Hanoi, Vietnam (N.T.T.H.); First Faculty of Medicine, Charles University, and General University Hospital, Prague, Czech Republic (M.Z.); AstraZeneca, Cambridge, United Kingdom (H.M.); AstraZeneca, Gaithersburg, MD (H.G., H.J.); and the Department of Radiation Oncology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam (S.S.)
| | - Helen Mann
- From Jilin Cancer Hospital, Changchun (Y. Cheng), Department II of Thoracic Oncology (J.F.) and the Department of Radiation Oncology (A.S.), Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, and the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou (Q.W.) - all in China; Sarah Cannon Research Institute (D.R.S.) and Tennessee Oncology (D.B.D.) - both in Nashville; Yonsei Cancer Center, Yonsei University College of Medicine, Seoul (B.C.C.), and Chungbuk National University Hospital, Cheongju (K.H.L.) - both in South Korea; Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology" of the Ministry of Health of the Russian Federation, Moscow (K.K.L.); Cancer and Hematology Centers of Western Michigan, Grand Rapids (Y. Chen); National Cancer Center Hospital East, Kashiwa (Y.Z.), and Kyushu University Hospital, Fukuoka (Y.S.) - both in Japan; Hospital Vall d'Hebron and Vall d'Hebron Institute of Oncology, Barcelona (A.N.), and Hospital Universitario Virgen del Rocio, Seville (R.B.) - both in Spain; Hospitals of the City of Cologne, Cologne, Germany (E.L.B.); Chang Gung Medical Foundation-Linkou Branch, Taoyuan, Taiwan (J.W.-C.C.); Akdeniz University Medical Faculty, Antalya, Turkey (S.S.G.); Medical University of Gdansk, Gdansk, Poland (A.B.); National Cancer Hospital, Hanoi, Vietnam (N.T.T.H.); First Faculty of Medicine, Charles University, and General University Hospital, Prague, Czech Republic (M.Z.); AstraZeneca, Cambridge, United Kingdom (H.M.); AstraZeneca, Gaithersburg, MD (H.G., H.J.); and the Department of Radiation Oncology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam (S.S.)
| | - Hema Gowda
- From Jilin Cancer Hospital, Changchun (Y. Cheng), Department II of Thoracic Oncology (J.F.) and the Department of Radiation Oncology (A.S.), Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, and the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou (Q.W.) - all in China; Sarah Cannon Research Institute (D.R.S.) and Tennessee Oncology (D.B.D.) - both in Nashville; Yonsei Cancer Center, Yonsei University College of Medicine, Seoul (B.C.C.), and Chungbuk National University Hospital, Cheongju (K.H.L.) - both in South Korea; Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology" of the Ministry of Health of the Russian Federation, Moscow (K.K.L.); Cancer and Hematology Centers of Western Michigan, Grand Rapids (Y. Chen); National Cancer Center Hospital East, Kashiwa (Y.Z.), and Kyushu University Hospital, Fukuoka (Y.S.) - both in Japan; Hospital Vall d'Hebron and Vall d'Hebron Institute of Oncology, Barcelona (A.N.), and Hospital Universitario Virgen del Rocio, Seville (R.B.) - both in Spain; Hospitals of the City of Cologne, Cologne, Germany (E.L.B.); Chang Gung Medical Foundation-Linkou Branch, Taoyuan, Taiwan (J.W.-C.C.); Akdeniz University Medical Faculty, Antalya, Turkey (S.S.G.); Medical University of Gdansk, Gdansk, Poland (A.B.); National Cancer Hospital, Hanoi, Vietnam (N.T.T.H.); First Faculty of Medicine, Charles University, and General University Hospital, Prague, Czech Republic (M.Z.); AstraZeneca, Cambridge, United Kingdom (H.M.); AstraZeneca, Gaithersburg, MD (H.G., H.J.); and the Department of Radiation Oncology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam (S.S.)
| | - Haiyi Jiang
- From Jilin Cancer Hospital, Changchun (Y. Cheng), Department II of Thoracic Oncology (J.F.) and the Department of Radiation Oncology (A.S.), Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, and the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou (Q.W.) - all in China; Sarah Cannon Research Institute (D.R.S.) and Tennessee Oncology (D.B.D.) - both in Nashville; Yonsei Cancer Center, Yonsei University College of Medicine, Seoul (B.C.C.), and Chungbuk National University Hospital, Cheongju (K.H.L.) - both in South Korea; Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology" of the Ministry of Health of the Russian Federation, Moscow (K.K.L.); Cancer and Hematology Centers of Western Michigan, Grand Rapids (Y. Chen); National Cancer Center Hospital East, Kashiwa (Y.Z.), and Kyushu University Hospital, Fukuoka (Y.S.) - both in Japan; Hospital Vall d'Hebron and Vall d'Hebron Institute of Oncology, Barcelona (A.N.), and Hospital Universitario Virgen del Rocio, Seville (R.B.) - both in Spain; Hospitals of the City of Cologne, Cologne, Germany (E.L.B.); Chang Gung Medical Foundation-Linkou Branch, Taoyuan, Taiwan (J.W.-C.C.); Akdeniz University Medical Faculty, Antalya, Turkey (S.S.G.); Medical University of Gdansk, Gdansk, Poland (A.B.); National Cancer Hospital, Hanoi, Vietnam (N.T.T.H.); First Faculty of Medicine, Charles University, and General University Hospital, Prague, Czech Republic (M.Z.); AstraZeneca, Cambridge, United Kingdom (H.M.); AstraZeneca, Gaithersburg, MD (H.G., H.J.); and the Department of Radiation Oncology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam (S.S.)
| | - Suresh Senan
- From Jilin Cancer Hospital, Changchun (Y. Cheng), Department II of Thoracic Oncology (J.F.) and the Department of Radiation Oncology (A.S.), Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, and the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou (Q.W.) - all in China; Sarah Cannon Research Institute (D.R.S.) and Tennessee Oncology (D.B.D.) - both in Nashville; Yonsei Cancer Center, Yonsei University College of Medicine, Seoul (B.C.C.), and Chungbuk National University Hospital, Cheongju (K.H.L.) - both in South Korea; Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology" of the Ministry of Health of the Russian Federation, Moscow (K.K.L.); Cancer and Hematology Centers of Western Michigan, Grand Rapids (Y. Chen); National Cancer Center Hospital East, Kashiwa (Y.Z.), and Kyushu University Hospital, Fukuoka (Y.S.) - both in Japan; Hospital Vall d'Hebron and Vall d'Hebron Institute of Oncology, Barcelona (A.N.), and Hospital Universitario Virgen del Rocio, Seville (R.B.) - both in Spain; Hospitals of the City of Cologne, Cologne, Germany (E.L.B.); Chang Gung Medical Foundation-Linkou Branch, Taoyuan, Taiwan (J.W.-C.C.); Akdeniz University Medical Faculty, Antalya, Turkey (S.S.G.); Medical University of Gdansk, Gdansk, Poland (A.B.); National Cancer Hospital, Hanoi, Vietnam (N.T.T.H.); First Faculty of Medicine, Charles University, and General University Hospital, Prague, Czech Republic (M.Z.); AstraZeneca, Cambridge, United Kingdom (H.M.); AstraZeneca, Gaithersburg, MD (H.G., H.J.); and the Department of Radiation Oncology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam (S.S.)
| |
Collapse
|
30
|
Cui Y, Chen Y, Zhao P, Li S, Cheng Y, Ren X. Peripheral NK cells identified as the predictor of response in extensive-stage small cell lung cancer patients treated with first-line immunotherapy plus chemotherapy. Clin Transl Oncol 2024; 26:2522-2530. [PMID: 38662170 DOI: 10.1007/s12094-024-03479-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 03/23/2024] [Indexed: 04/26/2024]
Abstract
PURPOSE Although immunotherapy improves outcomes in extensive-stage small-cell lung cancer (ES-SCLC), the search for biomarkers predicting treatment success is crucial. Natural killer (NK) cells are potential indicators in various cancers, however, their precise role in ES-SCLC prognosis remains unclear. METHODS In this retrospective study, 33 patients with ES-SCLC treated with first-line immuno-chemotherapy were enrolled. The peripheral NK cell percentage and its longitudinal dynamics were analyzed using flow cytometry. Progression-free survival (PFS) and overall survival (OS) were calculated as hazard ratio (HR) and compared statistically. RESULTS The median PFS was better in the group with normal baseline NK cell levels than the low group (7.0 vs. 4.6 months; HR = 0.17; 95% CI 0.07-0.41; P < 0.0001), but there was no association with OS (14.9 vs. 10.3 months; HR = 0.55; 95% CI 0.23-1.31; P = 0.171). Furthermore, the NK cell% for 95.0% of patients increased after immunochemotherapy in the clinical response group (P = 0.0047), which led to a better median PFS (6.3 vs. 2.1 months; HR = 0.23; 95% CI 0.05-0.98; P < 0.0001) and OS (14.9 vs. 5.9 months; HR = 0.20; 95% CI 0.04-1.02; P < 0.0001). Similar trends were observed with NK cell% changes up to disease progression, improving PFS (6.5 vs. 4.3; HR = 0.41; 95% CI 0.12-0.92; P = 0.0049) and OS (17.4 vs. 9.7; HR = 0.42; 95% CI 0.17-1.02; P < 0.0001). CONCLUSION In patients with ES-SCLC, the percentage and changes in peripheral NK cells can predict the response to combined immunotherapy and chemotherapy.
Collapse
Affiliation(s)
- Yanan Cui
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
- Division of Thoracic Oncology, Jilin Cancer Hospital, Changchun, China
| | - Yanping Chen
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Peiyan Zhao
- Translational Oncology Research Lab, Jilin Cancer Hospital, Changchun, China
| | - Shuang Li
- Big Data Center of Clinical, Jilin Cancer Hospital, Changchun, China
| | - Ying Cheng
- Division of Thoracic Oncology, Jilin Cancer Hospital, Changchun, China.
| | - Xiubao Ren
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China.
- Tianjin's Clinical Research Center for Cancer, Tianjin, China.
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China.
| |
Collapse
|
31
|
Reck M, Dziadziuszko R, Sugawara S, Kao S, Hochmair M, Huemer F, de Castro G, Havel L, Bernabé Caro R, Losonczy G, Lee JS, Kowalski DM, Andric Z, Califano R, Veatch A, Gerstner G, Batus M, Morris S, Kaul M, Cuchelkar V, Li H, Danner BJ, Nabet BY, Liu SV. Five-year survival in patients with extensive-stage small cell lung cancer treated with atezolizumab in the Phase III IMpower133 study and the Phase III IMbrella A extension study. Lung Cancer 2024; 196:107924. [PMID: 39306923 DOI: 10.1016/j.lungcan.2024.107924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/31/2024] [Accepted: 08/06/2024] [Indexed: 10/13/2024]
Abstract
OBJECTIVES IMbrella A is a Phase III extension study that allowed rollover from Roche/Genentech-sponsored atezolizumab trials, including IMpower133, a Phase I/III trial of first-line atezolizumab or placebo plus carboplatin/etoposide in extensive-stage small cell lung cancer. We report outcomes from an exploratory analysis of IMpower133 with extended time-to-event data for patients who rolled over to IMbrella A. MATERIALS AND METHODS IMpower133 patients could roll over to IMbrella A to receive atezolizumab 1200 mg intravenously every three weeks if they continued to receive atezolizumab at IMpower133 closure or were in survival follow-up after atezolizumab discontinuation. Overall survival and safety were assessed; only serious adverse events and AEs of special interest were collected in IMbrella A. RESULTS Eighteen of 26 eligible patients rolled over to IMbrella A. At clinical cutoff (March 16, 2023), median follow-up in the atezolizumab plus carboplatin/etoposide arm (IMpower133 and IMbrella A) was 59.4 months. The three-, four-, and five-year overall survival (95 % CI) estimates were 16 % (11 %-21 %), 13 % (8 %-18 %), and 12 % (7 %-17 %), respectively. In IMbrella A, serious adverse events occurred in three patients (16.7 %), and one adverse event of special interest was reported (grade two hypothyroidism). CONCLUSION This long-term analysis of patients from IMbrella A previously enrolled in IMpower133 provides the first report of five-year overall survival outcomes in patients with extensive-stage small cell lung cancer treated with first-line cancer immunotherapy and chemotherapy. While limited by small patient numbers and lack of long-term data for the IMpower133 control arm, exploratory overall survival analyses in patients treated with atezolizumab plus carboplatin/etoposide compared favorably with historical data with chemotherapy alone. NCT03148418.
Collapse
Affiliation(s)
- Martin Reck
- Lung Clinic Grosshansdorf, Airway Research Center North, German Center of Lung Research, Grosshansdorf, Germany.
| | - Rafal Dziadziuszko
- Faculty of Medicine, Department of Oncology and Radiotherapy, Medical University of Gdańsk, Gdańsk, Poland
| | - Shunichi Sugawara
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Steven Kao
- Department of Medical Oncology, Chris O'Brien Lifehouse, Camperdown, NSW, Australia
| | - Maximilian Hochmair
- Department of Respiratory and Critical Care Medicine, Karl Landsteiner Institute of Lung Research and Pulmonary Oncology, Vienna North Hospital Klinik Floridsdorf, Vienna, Austria
| | - Florian Huemer
- Department of Respiratory Care, Ludwig Boltzmann Institute for Lung Health, Klinik Penzing, Vienna, Austria
| | - Gilberto de Castro
- Clinical Oncology, Instituto de Cancer do Estado de São Paulo, Hospital Das Clínicas Da FMUSP, São Paulo, Brazil
| | - Libor Havel
- First Faculty of Medicine, Charles University, Thomayer Hospital, Prague, Czech Republic
| | - Reyes Bernabé Caro
- Department of Medical Oncology, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - György Losonczy
- Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Jong-Seok Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Dariusz M Kowalski
- Department of Lung Cancer and Chest Tumours, Maria Skłodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Zoran Andric
- Department of Medical Oncology, Clinical Hospital Centre Bezanijska Kosa, Belgrade, Serbia
| | - Raffaele Califano
- Department of Medical Oncology, The Christie NHS Foundation Trust and Division of Cancer Sciences, The University of Manchester, Manchester, UK
| | | | | | - Marta Batus
- Medical Oncology, Rush University Medical Center, Chicago, IL, USA
| | | | - Monika Kaul
- Oncology Product Development Safety, Genentech Inc, South San Francisco, CA, USA
| | - Vaikunth Cuchelkar
- Product Development Oncology-Hematology, Genentech, South San Francisco, CA, USA
| | - Huafei Li
- Roche, Product Development China, Shanghai, China
| | | | - Barzin Y Nabet
- Computational Sciences, Genentech, South San Francisco, CA, USA
| | - Stephen V Liu
- Thoracic Oncology and Developmental Therapeutics, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| |
Collapse
|
32
|
Akbulut D, Whiting K, Teo MY, Tallman JE, Ozcan GG, Basar M, Jia L, Rammal R, Chen JF, Sarungbam J, Chen YB, Gopalan A, Fine SW, Tickoo SK, Mehra R, Baine M, Bochner BH, Pietzak EJ, Bajorin DF, Rosenberg JE, Iyer G, Solit DB, Reuter VE, Rekhtman N, Ostrovnaya I, Al-Ahmadie H. Differential NEUROD1, ASCL1, and POU2F3 Expression Defines Molecular Subsets of Bladder Small Cell/Neuroendocrine Carcinoma With Prognostic Implications. Mod Pathol 2024; 37:100557. [PMID: 38964503 PMCID: PMC11490389 DOI: 10.1016/j.modpat.2024.100557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/22/2024] [Accepted: 06/18/2024] [Indexed: 07/06/2024]
Abstract
Small cell carcinomas (SMC) of the lung are now molecularly classified based on the expression of transcriptional regulators (NEUROD1, ASCL1, POU2F3, and YAP1) and DLL3, which has emerged as an investigational therapeutic target. PLCG2 has been shown to identify a distinct subpopulation of lung SMC with stem cell-like and prometastasis features and poor prognosis. We analyzed the expression of these novel neuroendocrine markers and their association with traditional neuroendocrine markers and patient outcomes in a cohort of bladder neuroendocrine carcinoma (NEC) consisting of 103 SMC and 19 large cell NEC (LCNEC) assembled in tissue microarrays. Coexpression patterns were assessed and integrated with detailed clinical annotation including overall (OS) and recurrence-free survival (RFS) and response to neoadjuvant/adjuvant chemotherapy. We identified 5 distinct molecular subtypes in bladder SMC based on the expression of ASCL1, NEUROD1, and POU2F3: ASCL1+/NEUROD1- (n = 33; 34%), ASCL1- /NEUROD1+ (n = 21; 21%), ASCL1+/NEUROD1+ (n = 17; 17%), POU2F3+ (n = 22, 22%), and ASCL1- /NEUROD1- /POU2F3- (n = 5, 5%). POU2F3+ tumors were mutually exclusive with those expressing ASCL1 and NEUROD1 and exhibited lower expression of traditional neuroendocrine markers. PLCG2 expression was noted in 33 tumors (32%) and was highly correlated with POU2F3 expression (P < .001). DLL3 expression was high in both SMC (n = 72, 82%) and LCNEC (n = 11, 85%). YAP1 expression was enriched in nonneuroendocrine components and negatively correlated with all neuroendocrine markers. In patients without metastatic disease who underwent radical cystectomy, PLCG2+ or POU2F3+ tumors had shorter RFS and OS (P < .05), but their expression was not associated with metastasis status or response to neoadjuvant/adjuvant chemotherapy. In conclusion, the NEC of the bladder can be divided into distinct molecular subtypes based on the expression of ASCL1, NEUROD1, and POU2F3. POU2F3-expressing tumors represent an ASCL1/NEUROD1-negative subset of bladder NEC characterized by lower expression of traditional neuroendocrine markers. Marker expression patterns were similar in SMC and LCNEC. Expression of PLCG2 and POU2F3 was associated with shorter RFS and OS. DLL3 was expressed at high levels in both SMC and LCNEC of the bladder, nominating it as a potential therapeutic target.
Collapse
MESH Headings
- Humans
- Basic Helix-Loop-Helix Transcription Factors/analysis
- Basic Helix-Loop-Helix Transcription Factors/metabolism
- Urinary Bladder Neoplasms/pathology
- Urinary Bladder Neoplasms/mortality
- Urinary Bladder Neoplasms/metabolism
- Biomarkers, Tumor/analysis
- Biomarkers, Tumor/metabolism
- Carcinoma, Neuroendocrine/pathology
- Carcinoma, Neuroendocrine/metabolism
- Carcinoma, Neuroendocrine/mortality
- Carcinoma, Neuroendocrine/therapy
- Male
- Female
- Aged
- Middle Aged
- Prognosis
- Carcinoma, Small Cell/pathology
- Carcinoma, Small Cell/metabolism
- Carcinoma, Small Cell/mortality
- Carcinoma, Small Cell/genetics
- Tissue Array Analysis
- POU Domain Factors/genetics
- POU Domain Factors/metabolism
- POU Domain Factors/analysis
- Adult
- Aged, 80 and over
- Immunohistochemistry
- Disease-Free Survival
Collapse
Affiliation(s)
- Dilara Akbulut
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Karissa Whiting
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Min-Yuen Teo
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jacob E Tallman
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Gamze Gokturk Ozcan
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Pathology and Laboratory Medicine, Henry Ford Hospital, Detroit, Michigan
| | - Merve Basar
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Liwei Jia
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Pathology, UT Southwestern, Dallas, Texas
| | - Rayan Rammal
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jie-Fu Chen
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Judy Sarungbam
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ying-Bei Chen
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Anuradha Gopalan
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Samson W Fine
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Satish K Tickoo
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Rohit Mehra
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Marina Baine
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Bernard H Bochner
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Eugene J Pietzak
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Dean F Bajorin
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jonathan E Rosenberg
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Gopa Iyer
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - David B Solit
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Victor E Reuter
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Natasha Rekhtman
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Irina Ostrovnaya
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Hikmat Al-Ahmadie
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
33
|
Jiang J, Han D, Wang J, Wen W, Zhang R, Qin W. Neuroendocrine transdifferentiation in human cancer: molecular mechanisms and therapeutic targets. MedComm (Beijing) 2024; 5:e761. [PMID: 39372390 PMCID: PMC11450264 DOI: 10.1002/mco2.761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/04/2024] [Accepted: 09/08/2024] [Indexed: 10/08/2024] Open
Abstract
Neuroendocrine transdifferentiation (NEtD), also commonly referred to as lineage plasticity, emerges as an acquired resistance mechanism to molecular targeted therapies in multiple cancer types, predominately occurs in metastatic epidermal growth factor receptor (EGFR)-mutant non-small cell lung cancer treated with EGFR tyrosine kinase inhibitors and metastatic castration-resistant prostate cancer treated with androgen receptor targeting therapies. NEtD tumors are the lethal cancer histologic subtype with unfavorable prognosis and limited treatment. A comprehensive understanding of molecular mechanism underlying targeted-induced plasticity could greatly facilitate the development of novel therapies. In the past few years, increasingly elegant studies indicated that NEtD tumors share key the convergent genomic and phenotypic characteristics irrespective of their site of origin, but also embrace distinct change and function of molecular mechanisms. In this review, we provide a comprehensive overview of the current understanding of molecular mechanism in regulating the NEtD, including genetic alterations, DNA methylation, histone modifications, dysregulated noncoding RNA, lineage-specific transcription factors regulation, and other proteomic alterations. We also provide the current management of targeted therapies in clinical and preclinical practice.
Collapse
Affiliation(s)
- Jun Jiang
- Department of UrologyXijing HospitalAir Force Medical UniversityXi'anChina
- Department of Health Service, Base of Health ServiceAir Force Medical UniversityXi'anChina
| | - Donghui Han
- Department of UrologyXijing HospitalAir Force Medical UniversityXi'anChina
| | - Jiawei Wang
- Department of Clinical Immunology, PLA Specialized Research Institute of Rheumatology & Immunology, Xijing Hospital, and National Translational Science Center for Molecular MedicineAir Force Medical UniversityXi'anChina
| | - Weihong Wen
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical ResearchNorthwestern Polytechnical UniversityXi'anChina
| | - Rui Zhang
- State Key Laboratory of Cancer BiologyDepartment of ImmunologyAir Force Medical UniversityXi'anChina
| | - Weijun Qin
- Department of UrologyXijing HospitalAir Force Medical UniversityXi'anChina
| |
Collapse
|
34
|
Redin E, Quintanal-Villalonga Á, Rudin CM. Small cell lung cancer profiling: an updated synthesis of subtypes, vulnerabilities, and plasticity. Trends Cancer 2024; 10:935-946. [PMID: 39164163 DOI: 10.1016/j.trecan.2024.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 07/16/2024] [Accepted: 07/25/2024] [Indexed: 08/22/2024]
Abstract
Small cell lung cancer (SCLC) is a devastating disease with high proliferative and metastatic capacity. SCLC has been classified into molecular subtypes based on differential expression of lineage-defining transcription factors. Recent studies have proposed new subtypes that are based on both tumor-intrinsic and -extrinsic factors. SCLC demonstrates substantial intratumoral subtype heterogeneity characterized by highly plastic transcriptional states, indicating that the initially dominant subtype can shift during disease progression and in association with resistance to therapy. Strategies to promote or constrain plasticity and cell fate transitions have nominated novel targets that could prompt the development of more durably effective therapies for patients with SCLC. In this review, we describe the latest advances in SCLC subtype classification and their biological and clinical implications.
Collapse
Affiliation(s)
- Esther Redin
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| | - Álvaro Quintanal-Villalonga
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Charles M Rudin
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Pharmacology Program, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
35
|
Maheshwari S, Gentzler RD. Implications for practice: phase II/III trial of carboplatin and irinotecan for elderly patients with extensive-stage small-cell lung cancer in Japan. J Thorac Dis 2024; 16:6342-6346. [PMID: 39444913 PMCID: PMC11494565 DOI: 10.21037/jtd-24-279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 09/10/2024] [Indexed: 10/25/2024]
Affiliation(s)
- Shipra Maheshwari
- Division of Hematology, Department of Medicine, University of Virginia Cancer Center, Charlottesville, VA, USA
| | - Ryan D Gentzler
- Division of Hematology, Department of Medicine, University of Virginia Cancer Center, Charlottesville, VA, USA
| |
Collapse
|
36
|
Hong Q, Liang S, Han R, Zhao C, Mu J. Signature stemmed from two transcription factor families determines histological fate and regulates immune infiltration in patients with lung cancer. J Thorac Dis 2024; 16:5663-5674. [PMID: 39444905 PMCID: PMC11494554 DOI: 10.21037/jtd-24-733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 07/26/2024] [Indexed: 10/25/2024]
Abstract
Background Earlier research has reported that transcription factors play a crucial role in the anti-tumorigenic immune response of lung cancer patients. The aim of this study is to determine the relationship between post-translational modifications of transcription factors and histological fate and patient prognosis. Methods Based on the information of 293 lung cancer patients in the Gene Expression Omnibus (GEO) database, differentially expressed genes (DEGs) related to the interferon regulatory factor (IRF) and signal transducer and activator of transcription (STAT) families between patients experiencing early death and those with long-term survival were identified and characterized. A survival prediction model was established by incorporating 7 STAT genes and 9 IRF genes into the least absolute shrinkage and selection operator (LASSO) algorithm. Gene Ontology (GO) enrichment analysis indicated that these two transcription factor families can govern lung cancer tissue differentiation and predict patient prognosis. Moreover, the Cox proportional hazards regression model was applied to select the genes with the highest predictive capability to construct a gene-based signature. Lastly, the data of 1,803 and 784 lung cancer patients from the Kaplan-Meier plotter (KMPLOT) and The Cancer Genome Atlas (TCGA) databases were used to evaluate the accuracy and sensitivity of the model. Results Based on the minimum criterion, TRIM28, IRF3, and STAT3 were employed to generate the prognostic model. The 1-, 3-, and 5-year area under the curve (AUC) values of the three-gene-based signature showed consistent results, signifying that the model had excellent accuracy and sensitivity in predicting overall survival (OS) for patients with lung cancer. Finally, the three-gene signature and tumor-node-metastasis (TNM) staging system were combined to construct a nomogram for evaluating the OS of lung cancer patients. TRIM28 may affect the stability of IRF3. Encouragingly, the predicted OS was highly consistent with the observed OS in multiple cohorts. Conclusions Taken together, these findings implied that the predictive model based on the three-gene signature showed robust discriminatory performance.
Collapse
Affiliation(s)
- Qian Hong
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shuang Liang
- State Key Laboratory of Molecular Oncology, Department of Clinical Laboratory, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rui Han
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chenguang Zhao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Juwei Mu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
37
|
Liu T, Chen X, Mo S, Zhou T, Ma W, Chen G, Chen X, Shi M, Yang Y, Huang Y, Zhao H, Fang W, Yang Y, Li J, Zhang L, Zhao Y. Molecular subtypes and prognostic factors of lung large cell neuroendocrine carcinoma. Transl Lung Cancer Res 2024; 13:2222-2235. [PMID: 39430332 PMCID: PMC11484736 DOI: 10.21037/tlcr-24-292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 07/26/2024] [Indexed: 10/22/2024]
Abstract
Background Lung large cell neuroendocrine carcinoma (LCNEC) is an aggressive disease with poor prognosis and short-term survival, which lacks effective prognostic indicators. The study aims to investigate the molecular subtypes and prognostic markers of lung LCNEC. Methods Patients diagnosed with lung LCNEC at Sun Yat-sen University Cancer Center (SYSUCC) between November 2007 and January 2021 were screened. Baseline clinical data were collected and routine blood indexes including lymphocyte-to-monocyte ratio (LMR), neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR) and systemic immune-inflammation index (SII) were calculated. Immunohistochemistry (IHC) of ASCL1, NEUROD1, POU2F3, YAP1 were done to perform molecular subtyping, while CD56, Syn, CgA, CD3, CD8, CD20, CD68, and CD163 were also stained on tissue samples. Then prognostic factors of lung LCNEC were explored. Results One hundred and fifty-one lung LCNEC patients were identified, 103 of whom had complete clinical information, available routine blood and biochemical indexes were eventually included in the present study. Tumor tissue specimens were available from 64 patients. Positive expression rates of ASCL1, NEUROD1, and YAP1 were 82.8%, 50.0%, and 28.1%, respectively. No POU2F3+ cases were detected. Forty (62.5%) patients co-expressed with two or three markers. High LMR (>3.3) was an independent predictor of favorable prognosis of disease-free survival (DFS) [hazard ratio (HR), 0.391; 95% confidence interval (CI): 0.161-0.948; P=0.04] and overall survival (OS) (HR, 0.201; 95% CI: 0.071-0.574; P=0.003). Notably, high LMR was correlated with higher intra-tumoral CD3+ (P=0.004), CD8+ (P=0.01), and CD68+ (P<0.001) immune cell infiltration compared to low LMR in lung LCNEC. Conclusions Our study validated molecular subtypes by IHC in lung LCNEC, and co-expression was found among different subtypes, with no prognostic effect. High blood LMR level was associated with a favorable prognosis in lung LCNEC, which might partly reflect a hot tumor tissue immune microenvironment. Our findings may benefit clinical practice, and further studies are warranted.
Collapse
Affiliation(s)
- Tingting Liu
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xueyuan Chen
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Silang Mo
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ting Zhou
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wenjuan Ma
- Department of Intensive Care Unit, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Gang Chen
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiang Chen
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Mengting Shi
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yuwen Yang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yan Huang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hongyun Zhao
- Department of Clinical Research, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wenfeng Fang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yunpeng Yang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jing Li
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Li Zhang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yuanyuan Zhao
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
38
|
Kaczorowski M, Ylaya K, Chłopek M, Lasota J, Miettinen M. Expression of POU2F3 Transcription Factor and POU2AF2, POU2F3 Coactivator, in Tuft Cell-like Carcinoma and Other Tumors. Am J Surg Pathol 2024:00000478-990000000-00419. [PMID: 39319626 DOI: 10.1097/pas.0000000000002313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
Abstract
Epithelial chemosensory cells in hollow organs, also known as tuft cells, were implicated in tumorigenesis, including a tuft cell-like small cell lung carcinoma. Expression of the POU2F3 transcription factor is a marker of tuft cell lineage. However, tuft cell development, differentiation, and proliferation are controlled by the expression of the complex formed by POU2F3 and POU2AF2 or POU2AF3 transcriptional coactivators. A cohort of epithelial (n=6064) and mesenchymal/neuroectodermal (n=2730) tumors was screened for POU2F3 expression by immunohistochemistry. Variable immunoreactivity ranging from diffuse to scattered positive cells was found in ∼12.4% of epithelial and 4.6% of mesenchymal/neuroectodermal tumors. Cases with predominantly diffuse or patchy POU2F3 positivity representing various types of malignant tumors (n=43) were selected for further study, including POU2AF2 immunohistochemistry. Thirteen of 15 tumors with neuroendocrine differentiation originating from the lung, colon, head and neck, skin, and bladder revealed diffuse POU2F3 positivity. Most of those tumors (n=9) co-expressed POU2AF2, usually extensively. Seven squamous and basal cell carcinomas from the oral cavity, skin, lung, and thymus with diffuse POU2F3 immunostaining except one, lacked POU2AF2 expression. Other variably POU2F3-positive carcinomas (n=13) from the colon, pancreas, liver, kidney, testis, endometrium, ovary, and breast lacked POU2AF2 immunoreactivity. All POU2F3-positive mesenchymal and neuroectodermal tumors (n=8), including synovial sarcoma, solitary fibrous tumor, glioblastoma, Wilms tumor, and melanoma were POU2AF2-negative. POU2F3 expression is a highly sensitive but nonspecific indicator of tuft cell differentiation. Co-expression of POU2F3 and POU2AF2 appears to be a more specific marker, although it may not pinpoint tumors driven by the POU2F3-POU2AF3 complex.
Collapse
Affiliation(s)
- Maciej Kaczorowski
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD
- Department of Clinical and Experimental Pathology, Wroclaw Medical University, Wrocław, Poland
| | - Kris Ylaya
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD
| | | | - Jerzy Lasota
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD
| | - Markku Miettinen
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD
| |
Collapse
|
39
|
Reinmuth N, Juan-Vidal O, Kowalski D, Bryl M, Kryzhanivska A, Vicente D, Horváth Z, Gálffy G, Csánky E, Pápai Székely Z, Vynnychenko I, Armstrong J, Dalvi T, Xie M, Iyer S, Shrestha Y, Jiang H, Bondarenko I. Novel Combinations of Immunotherapies or DNA Damage Repair Inhibitors in Platinum-Refractory Extensive-Stage Small Cell Lung Cancer: The Phase II BALTIC Study. Clin Cancer Res 2024; 30:4055-4067. [PMID: 39017667 PMCID: PMC11393542 DOI: 10.1158/1078-0432.ccr-24-0013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 04/12/2024] [Accepted: 07/16/2024] [Indexed: 07/18/2024]
Abstract
PURPOSE The phase II, multiarm, signal-searching BALTIC study (NCT02937818) assessed novel treatment combinations for platinum-refractory/resistant extensive-stage small cell lung cancer (ES-SCLC). PATIENTS AND METHODS Patients with ES-SCLC with progressive disease during or within 90 days of completing first-line platinum-based chemotherapy received one of three regimens: durvalumab plus tremelimumab followed by durvalumab monotherapy (arm A), adavosertib plus carboplatin (arm B), or ceralasertib plus olaparib (arm C). The primary endpoint was the objective response rate. Prespecified exploratory biomarker analyses were conducted in arms A and C. RESULTS In arm A (n = 41), arm B (n = 10), and arm C (n = 21), the confirmed objective response rates were 7.3%, 0%, and 4.8%, respectively. Safety profiles in all arms were consistent with those of the individual drugs. In arm A, patients with PD-L1 expression (tumor cells or immune cells) ≥1% seemed to have a greater likelihood of achieving disease control with durvalumab plus tremelimumab than those with PD-L1 (tumor cells and immune cells) <1%, and lower baseline ctDNA and reduction in the on-treatment ctDNA level were both associated with longer overall survival. Among patients treated with ceralasertib plus olaparib in arm C, specific immune response-relevant circulating chemokines and cytokines were identified as early biomarkers of survival and pharmacodynamic biomarkers. CONCLUSIONS In BALTIC, all combination regimens demonstrated tolerable safety profiles, but antitumor activity was limited in refractory/resistant ES-SCLC. Among patients treated with durvalumab plus tremelimumab, an association of on-treatment reduction in ctDNA with longer overall survival suggests the potential use of ctDNA as a surrogate of treatment response, warranting further investigation.
Collapse
Affiliation(s)
- Niels Reinmuth
- Department of Thoracic Oncology, Asklepios Lung Clinic Munich-Gauting, Gauting, Germany.
- German Center of Lung Research, LMU Munich, Munich, Germany.
| | - Oscar Juan-Vidal
- Department of Medical Oncology, La Fe University Hospital, Valencia, Spain.
| | - Dariusz Kowalski
- Department of Lung Cancer and Thoracic Tumours, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland.
| | - Maciej Bryl
- Oncology Department, E.J. Zeyland Wielkopolska Center of Pulmonology and Thoracic Surgery, Poznan, Poland.
| | - Anna Kryzhanivska
- Ivano-Frankivsk National Medical University, Ivano-Frankivsk, Ukraine.
| | - David Vicente
- Hospital Universitario Virgen Macarena, Seville, Spain.
| | - Zsolt Horváth
- Bacs-Kiskun County Teaching Hospital, Kecskemét, Hungary.
| | | | - Eszter Csánky
- Department of Pulmonology, Semmelweis Hospital, Miskolc, Hungary.
| | | | | | | | | | | | | | | | | | - Igor Bondarenko
- Dnipropetrovsk State Medical Academy, Dnipropetrovsk, Ukraine.
| |
Collapse
|
40
|
Chen T, Wang M, Chen Y, Cao Y, Liu Y. Advances in predictive biomarkers associated with immunotherapy in extensive-stage small cell lung cancer. Cell Biosci 2024; 14:117. [PMID: 39267195 PMCID: PMC11391723 DOI: 10.1186/s13578-024-01283-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 08/05/2024] [Indexed: 09/14/2024] Open
Abstract
Small cell lung cancer (SCLC) is a highly malignant and poor-prognosis cancer, with most cases diagnosed at the extensive stage (ES). Amidst a landscape marked by limited progress in treatment modalities for ES-SCLC over the past few decades, the integration of immune checkpoint inhibitors (ICIs) with platinum-based chemotherapy has provided a milestone approach for improving prognosis, emerging as the new standard for initial therapy in ES-SCLC. However, only a minority of SCLC patients can benefit from ICIs, which frequently come with varying degrees of immune-related adverse events (irAEs). Therefore, it is crucial to investigate predictive biomarkers to screen potential beneficiaries of ICIs, mitigate the risk of side effects, and improve treatment precision. This review summarized potential biomarkers for predicting ICI response in ES-SCLC, with a primary focus on markers sourced from tumor tissue or peripheral blood samples. The former mainly included PD-L1 expression, tumor mutational burden (TMB), along with cellular or molecular components related to the tumor microenvironment (TME) and antigen presentation machinery (APM), molecular subtypes of SCLC, and inflammatory gene expression profiles. Circulating biomarkers predominantly comprised circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), cytokines, plasma autoantibodies, inflammation-related parameters, and blood TMB. We synthesized and analyzed the research progress of these potential markers. Notably, investigations into PD-L1 expression and TMB have been the most extensive, exhibiting preliminary predictive efficacy in salvage immunotherapy; however, consistent conclusions have yet to be reached across studies. Additionally, novel predictive markers developed based on TME composition, APM, transcriptomic and genomic features provide promising tools for precision immunotherapy. Circulating biomarkers offer the advantages of convenience, non-invasiveness, and a comprehensive reflection of tumor molecular characteristics. They may serve as alternative options for predicting immunotherapy efficacy in SCLC. However, there is a scarcity of studies, and the significant heterogeneity in research findings warrants attention.
Collapse
Affiliation(s)
- Tong Chen
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Mingzhao Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Yanchao Chen
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Yang Cao
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Yutao Liu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China.
| |
Collapse
|
41
|
Li X, Tong L, Wang S, Yu J, Lu B, Wang Q, Hu M, Wu J, Yu J, Li B, Zhang T. Integration of clinical and blood parameters in risk prognostication for patients receiving immunochemotherapy for extensive stage small cell lung cancer: real-world data from two centers. BMC Med 2024; 22:381. [PMID: 39256789 PMCID: PMC11389556 DOI: 10.1186/s12916-024-03612-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 09/04/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) had modest advances in the treatment of extensive-stage small cell lung cancer (ES-SCLC) in clinical trials, but there is a lack of biomarkers for prognosis in clinical practice. METHODS We retrospectively collected data from ES-SCLC patients who received ICIs combined chemotherapy from two centers in China, integrated clinical and blood parameters, and constructed risk prognostication for immunochemotherapy. The population was divided into high- and low-risk groups, and the performance of the model was assessed separately in the training and validation cohorts. RESULTS Two hundred and twenty and 43 patients were included in the training and validation groups, respectively. The important predictors were screened including body mass index, liver metastases, coefficient variation of red blood cell distribution width, lactate dehydrogenase, albumin, and C-reactive protein. Predicting 1-year overall survival (OS), the AUC values under ROC for the model under training, internal validation, and external validation were 0.760, 0.732, and 0.722, respectively, and the calibration curve and clinical decision curve performed well. Applied the model to divide patients into low-risk and high-risk groups, and the median OS was 23.7 months and 9.1 months, and the median progression-free survival was 8.2 months and 4.8 months, respectively; furthermore, this ability to discriminate survival was also observed in the validation cohort. CONCLUSIONS We constructed a novel prognostic model for ES-SCLC to predict survival employing baseline tumor burden, nutritional and inflammatory parameters, it is easily measured to screen high-risk patient populations.
Collapse
Affiliation(s)
- Xiaomi Li
- Department of Oncology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
- Department of Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Li Tong
- Department of Oncology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Shan Wang
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Jiaqi Yu
- First Clinical Medical College, Shaanxi University of Traditional Chinese Medicine, Xianyang, 712000, Shaanxi, China
| | - Baohua Lu
- Department of Oncology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Qunhui Wang
- Department of Oncology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Mingming Hu
- Department of Oncology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Jinxiang Wu
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Jing Yu
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China.
| | - Baolan Li
- Department of Oncology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China.
| | - Tongmei Zhang
- Department of Oncology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China.
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
42
|
Hartmann GG, Sage J. Small Cell Lung Cancer Neuronal Features and Their Implications for Tumor Progression, Metastasis, and Therapy. Mol Cancer Res 2024; 22:787-795. [PMID: 38912893 PMCID: PMC11374474 DOI: 10.1158/1541-7786.mcr-24-0265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/30/2024] [Accepted: 06/20/2024] [Indexed: 06/25/2024]
Abstract
Small cell lung cancer (SCLC) is an epithelial neuroendocrine form of lung cancer for which survival rates remain dismal and new therapeutic approaches are greatly needed. Key biological features of SCLC tumors include fast growth and widespread metastasis, as well as rapid resistance to treatment. Similar to pulmonary neuroendocrine cells, SCLC cells have traits of both hormone-producing cells and neurons. In this study, we specifically discuss the neuronal features of SCLC. We consider how neuronal G protein-coupled receptors and other neuronal molecules on the surface of SCLC cells can contribute to the growth of SCLC tumors and serve as therapeutic targets in SCLC. We also review recent evidence for the role of neuronal programs expressed by SCLC cells in the fast proliferation, migration, and metastasis of these cells. We further highlight how these neuronal programs may be particularly relevant for the development of brain metastases and how they can assist SCLC cells to functionally interact with neurons and astrocytes. A greater understanding of the molecular and cellular neuronal features of SCLC is likely to uncover new vulnerabilities in SCLC cells, which may help develop novel therapeutic approaches. More generally, the epithelial-to-neuronal transition observed during tumor progression in SCLC and other cancer types can contribute significantly to tumor development and response to therapy.
Collapse
Affiliation(s)
- Griffin G. Hartmann
- Departments of Pediatrics and Genetics, Stanford University, Stanford, CA, USA
| | - Julien Sage
- Departments of Pediatrics and Genetics, Stanford University, Stanford, CA, USA
| |
Collapse
|
43
|
Jin Y, Wu Y, Reuben A, Zhu L, Gay CM, Wu Q, Zhou X, Mo H, Zheng Q, Ren J, Fang Z, Peng T, Wang N, Ma L, Fan Y, Song H, Zhang J, Chen M. Single-cell and spatial proteo-transcriptomic profiling reveals immune infiltration heterogeneity associated with neuroendocrine features in small cell lung cancer. Cell Discov 2024; 10:93. [PMID: 39231924 PMCID: PMC11375181 DOI: 10.1038/s41421-024-00703-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 06/23/2024] [Indexed: 09/06/2024] Open
Abstract
Small cell lung cancer (SCLC) is an aggressive pulmonary neuroendocrine malignancy featured by cold tumor immune microenvironment (TIME), limited benefit from immunotherapy, and poor survival. The spatial heterogeneity of TIME significantly associated with anti-tumor immunity has not been systemically studied in SCLC. We performed ultra-high-plex Digital Spatial Profiling on 132 tissue microarray cores from 44 treatment-naive limited-stage SCLC tumors. Incorporating single-cell RNA-sequencing data from a local cohort and published SCLC data, we established a spatial proteo-transcriptomic landscape covering over 18,000 genes and 60 key immuno-oncology proteins that participate in signaling pathways affecting tumorigenesis, immune regulation, and cancer metabolism across 3 pathologically defined spatial compartments (pan-CK-positive tumor nest; CD45/CD3-positive tumor stroma; para-tumor). Our study depicted the spatial transcriptomic and proteomic TIME architecture of SCLC, indicating clear intra-tumor heterogeneity dictated via canonical neuroendocrine subtyping markers; revealed the enrichment of innate immune cells and functionally impaired B cells in tumor nest and suggested potentially important immunoregulatory roles of monocytes/macrophages. We identified RE1 silencing factor (REST) as a potential biomarker for SCLC associated with low neuroendocrine features, more active anti-tumor immunity, and prolonged survival.
Collapse
Affiliation(s)
- Ying Jin
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang, China
| | - Yuefeng Wu
- The MOE Key Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Department of Cardiovascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE), School of Medicine, Zhejiang University, Haining, Zhejiang, China
| | - Alexandre Reuben
- Department of Thoracic/Head and Neck Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Liang Zhu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Carl M Gay
- Department of Thoracic/Head and Neck Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Qingzhe Wu
- The MOE Key Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xintong Zhou
- The MOE Key Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
| | - Haomin Mo
- The MOE Key Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
| | - Qi Zheng
- Department of Cardiovascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Junyu Ren
- College of Information Science and Electronic Engineering, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhaoyuan Fang
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE), School of Medicine, Zhejiang University, Haining, Zhejiang, China
| | - Teng Peng
- Department of Cardiovascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Nan Wang
- Cosmos Wisdom Biotech Co. Ltd., Hangzhou, Zhejiang, China
| | - Liang Ma
- Department of Cardiovascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yun Fan
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China.
| | - Hai Song
- The MOE Key Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China.
- Center for Oncology Medicine, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, Zhejiang, China.
| | - Jianjun Zhang
- Department of Thoracic/Head and Neck Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Ming Chen
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China.
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China.
- United Laboratory of Frontier Radiotherapy Technology of Sun Yat-sen University & Chinese Academy of Sciences Ion Medical Technology Co., Ltd, Guangzhou, Guangdong, China.
| |
Collapse
|
44
|
Choudhury NJ, Lai WV, Makhnin A, Heller G, Eng J, Li B, Preeshagul I, Santini FC, Offin M, Ng K, Paik P, Larsen C, Ginsberg MS, Lau Y, Zhang X, Baine MK, Rekhtman N, Rudin CM. A Phase I/II Study of Valemetostat (DS-3201b), an EZH1/2 Inhibitor, in Combination with Irinotecan in Patients with Recurrent Small-Cell Lung Cancer. Clin Cancer Res 2024; 30:3697-3703. [PMID: 38940666 PMCID: PMC11371507 DOI: 10.1158/1078-0432.ccr-23-3383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 02/12/2024] [Accepted: 06/26/2024] [Indexed: 06/29/2024]
Abstract
PURPOSE Recurrent small-cell lung cancer (SCLC) has few effective treatments. The EZH2-SLFN11 pathway is a driver of acquired chemoresistance that may be targeted. PATIENTS AND METHODS This phase I/II trial investigated valemetostat, an EZH1/2 inhibitor, with fixed-dose irinotecan in patients with recurrent SCLC. Phase I primary objectives were to assess safety, tolerability, and a recommended phase II dose (RP2D). The phase II primary objective was overall response rate (ORR), with secondary objectives of determining duration of response (DoR), progression-free survival (PFS), and overall survival (OS). Correlative analyses included immunohistochemistry of pretreatment and on-treatment tumor biopsies and pharmacokinetics analysis. RESULTS Twenty-two patients were enrolled (phase I, n = 12; phase II, n = 10); one withdrew consent prior to treatment. Three dose-limiting toxicities (DLT) in dose-escalation resulted in valemetostat 100 mg orally daily selected as RP2D. Among 21 evaluable patients, the most frequent (≥20%) treatment-related adverse events were diarrhea, fatigue, nausea, and rash; three patients discontinued treatment for toxicity. Three of the first 10 patients in phase II experienced DLTs triggering a stopping rule. The ORR was 4/19 or 21% [95% confidence interval (CI), 6%-46%]. The median DoR, PFS, and OS were 4.6 months, 2.2 months (95% CI, 1.3-7.6 months), and 6.6 months (95% CI, 4.3 to not reached), respectively. SLFN11/EZH2 expression and SCLC subtyping markers did not correlate with response, but MHC-I expression did increase with treatment. Two responders demonstrated subtype switching on treatment. CONCLUSIONS Combination valemetostat and irinotecan was not tolerated but demonstrated efficacy in recurrent SCLC. Valemetostat, combined with agents without overlapping toxicity, warrants further investigation in SCLC.
Collapse
Affiliation(s)
- Noura J Choudhury
- Thoracic Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - W Victoria Lai
- Thoracic Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alex Makhnin
- Thoracic Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Glenn Heller
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Juliana Eng
- Thoracic Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Bob Li
- Thoracic Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Isabel Preeshagul
- Thoracic Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Fernando C Santini
- Thoracic Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael Offin
- Thoracic Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Kenneth Ng
- Thoracic Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Paul Paik
- Thoracic Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Christina Larsen
- Thoracic Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michelle S Ginsberg
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Yvonne Lau
- Daiichi Sankyo, Inc., Basking Ridge, New Jersey
| | | | - Marina K Baine
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Natasha Rekhtman
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Charles M Rudin
- Thoracic Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| |
Collapse
|
45
|
Behrouzi R, Clipson A, Simpson KL, Blackhall F, Rothwell DG, Dive C, Mouliere F. Cell-free and extrachromosomal DNA profiling of small cell lung cancer. Trends Mol Med 2024:S1471-4914(24)00218-1. [PMID: 39232927 DOI: 10.1016/j.molmed.2024.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 09/06/2024]
Abstract
Small cell lung cancer (SCLC) is highly aggressive with poor prognosis. Despite a relative prevalence of circulating tumour DNA (ctDNA) in SCLC, liquid biopsies are not currently implemented, unlike non-SCLC where cell-free DNA (cfDNA) mutation profiling in the blood has utility for guiding targeted therapies and assessing minimal residual disease. cfDNA methylation profiling is highly sensitive for SCLC detection and holds promise for disease monitoring and molecular subtyping; cfDNA fragmentation profiling has also demonstrated clinical potential. Extrachromosomal DNA (ecDNA), that is often observed in SCLC, promotes tumour heterogeneity and chemotherapy resistance and can be detected in blood. We discuss how these cfDNA profiling modalities can be harnessed to expand the clinical applications of liquid biopsy in SCLC.
Collapse
Affiliation(s)
- Roya Behrouzi
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK; Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, UK; Cancer Research UK National Biomarker Centre, University of Manchester, Manchester, UK; Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK
| | - Alexandra Clipson
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester, UK
| | - Kathryn L Simpson
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester, UK; Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK
| | - Fiona Blackhall
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK; Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, UK
| | - Dominic G Rothwell
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester, UK
| | - Caroline Dive
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester, UK; Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK
| | - Florent Mouliere
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester, UK.
| |
Collapse
|
46
|
El Zarif T, Meador CB, Qiu X, Seo JH, Davidsohn MP, Savignano H, Lakshminarayanan G, McClure HM, Canniff J, Fortunato B, Li R, Banwait MK, Semaan K, Eid M, Long H, Hung YP, Mahadevan NR, Barbie DA, Oser MG, Piotrowska Z, Choueiri TK, Baca SC, Hata AN, Freedman ML, Berchuck JE. Detecting Small Cell Transformation in Patients with Advanced EGFR Mutant Lung Adenocarcinoma through Epigenomic cfDNA Profiling. Clin Cancer Res 2024; 30:3798-3811. [PMID: 38912901 PMCID: PMC11369616 DOI: 10.1158/1078-0432.ccr-24-0466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 04/04/2024] [Accepted: 06/20/2024] [Indexed: 06/25/2024]
Abstract
PURPOSE Histologic transformation to small cell lung cancer (SCLC) is a mechanism of treatment resistance in patients with advanced oncogene-driven lung adenocarcinoma (LUAD) that currently requires histologic review for diagnosis. Herein, we sought to develop an epigenomic cell-free DNA (cfDNA)-based approach to noninvasively detect small cell transformation in patients with EGFR mutant (EGFRm) LUAD. EXPERIMENTAL DESIGN To characterize the epigenomic landscape of transformed (t)SCLC relative to LUAD and de novo SCLC, we performed chromatin immunoprecipitation sequencing (ChIP-seq) to profile the histone modifications H3K27ac, H3K4me3, and H3K27me3; methylated DNA immunoprecipitation sequencing (MeDIP-seq); assay for transposase-accessible chromatin sequencing; and RNA sequencing on 26 lung cancer patient-derived xenograft (PDX) tumors. We then generated and analyzed H3K27ac ChIP-seq, MeDIP-seq, and whole genome sequencing cfDNA data from 1 mL aliquots of plasma from patients with EGFRm LUAD with or without tSCLC. RESULTS Analysis of 126 epigenomic libraries from the lung cancer PDXs revealed widespread epigenomic reprogramming between LUAD and tSCLC, with a large number of differential H3K27ac (n = 24,424), DNA methylation (n = 3,298), and chromatin accessibility (n = 16,352) sites between the two histologies. Tumor-informed analysis of each of these three epigenomic features in cfDNA resulted in accurate noninvasive discrimination between patients with EGFRm LUAD versus tSCLC [area under the receiver operating characteristic curve (AUROC) = 0.82-0.87]. A multianalyte cfDNA-based classifier integrating these three epigenomic features discriminated between EGFRm LUAD versus tSCLC with an AUROC of 0.94. CONCLUSIONS These data demonstrate the feasibility of detecting small cell transformation in patients with EGFRm LUAD through epigenomic cfDNA profiling of 1 mL of patient plasma.
Collapse
Affiliation(s)
- Talal El Zarif
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Department of Medicine, Yale School of Medicine, New Haven, Connecticut.
| | - Catherine B. Meador
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts.
| | - Xintao Qiu
- Department of Medicine, Yale School of Medicine, New Haven, Connecticut.
| | - Ji-Heui Seo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Matthew P. Davidsohn
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Hunter Savignano
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Gitanjali Lakshminarayanan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Heather M. McClure
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - John Canniff
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Brad Fortunato
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Rong Li
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Mandeep K. Banwait
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts.
| | - Karl Semaan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Eli and Edythe L. Broad Institute, Cambridge, Massachusetts.
| | - Marc Eid
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Henry Long
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Yin P. Hung
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Navin R. Mahadevan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts.
| | - David A. Barbie
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Matthew G. Oser
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Zofia Piotrowska
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts.
| | - Toni K. Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Sylvan C. Baca
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Eli and Edythe L. Broad Institute, Cambridge, Massachusetts.
| | - Aaron N. Hata
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts.
| | - Matthew L. Freedman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Eli and Edythe L. Broad Institute, Cambridge, Massachusetts.
| | - Jacob E. Berchuck
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| |
Collapse
|
47
|
Matsumoto N, Tajima K, Takahashi F, Mitsuishi Y, Wirawan A, Hidayat M, Winardi W, Wibowo A, Hayakawa D, Izumi K, Kanamori K, Miyashita Y, Handa T, Asao T, Ko R, Shukuya T, Shimada N, Takamochi K, Hayashi T, Suzuki K, Takahashi K. Clinical potential of SKP2 as diagnostic marker and therapeutic target in small cell lung cancer. Respir Investig 2024; 62:901-909. [PMID: 39116798 DOI: 10.1016/j.resinv.2024.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 06/27/2024] [Accepted: 07/22/2024] [Indexed: 08/10/2024]
Abstract
BACKGROUND Small cell lung cancer (SCLC) is the most aggressive type of lung cancer. The overall survival has not improved significantly over the last decades because no major therapeutic breakthroughs have been achieved for over 15 years. METHODS We analyzed a genome-wide loss-of-function screening database to identify vulnerabilities in SCLC for the development of urgently needed novel therapies. RESULTS We identified SKP2 (encoding S-phase kinase-associated protein 2) and CKS1B (encoding CDC28 protein kinase regulatory subunit 1B) as the two most essential genes in that order in SCLC. Notably, SKP2 and CKS1B comprise the p27 binding pocket of the E3 ubiquitin ligase SCFSKP2 complex. Immunohistochemistry on tissue microarrays revealed that SKP2 was expressed in >95% of samples at substantially higher levels than that observed for commonly used neuroendocrine markers. As expected, SCLC cell lines were sensitive to SKP2 inhibition. Furthermore, SKP2 or CKS1B knockdown induced apoptosis in RB1 mutant cells, whereas it induced senescence in RB1 wild-type cells. CONCLUSION Although the mechanism underlying SKP2 knockdown-induced growth inhibition differs between RB1-wild-type and -mutant SCLC, SKP2 can be considered a novel therapeutic target for patients with SCLC regardless of the RB1 mutation status. Our findings indicate that SKP2 is a potential novel clinical diagnostic marker and therapeutic target in SCLC.
Collapse
Affiliation(s)
- Naohisa Matsumoto
- Department of Respiratory Medicine, Juntendo University, Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan; Research Institute for Diseases of Old Ages, Juntendo University, Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
| | - Ken Tajima
- Department of Respiratory Medicine, Juntendo University, Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan; Research Institute for Diseases of Old Ages, Juntendo University, Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan.
| | - Fumiyuki Takahashi
- Department of Respiratory Medicine, Juntendo University, Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan; Research Institute for Diseases of Old Ages, Juntendo University, Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
| | - Yoichiro Mitsuishi
- Department of Respiratory Medicine, Juntendo University, Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan; Research Institute for Diseases of Old Ages, Juntendo University, Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
| | - Aditya Wirawan
- Department of Respiratory Medicine, Juntendo University, Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan; Research Institute for Diseases of Old Ages, Juntendo University, Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
| | - Moulid Hidayat
- Department of Respiratory Medicine, Juntendo University, Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan; Research Institute for Diseases of Old Ages, Juntendo University, Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
| | - Wira Winardi
- Department of Respiratory Medicine, Juntendo University, Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan; Research Institute for Diseases of Old Ages, Juntendo University, Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
| | - Adityo Wibowo
- Department of Respiratory Medicine, Juntendo University, Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan; Research Institute for Diseases of Old Ages, Juntendo University, Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
| | - Daisuke Hayakawa
- Department of Respiratory Medicine, Juntendo University, Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan; Research Institute for Diseases of Old Ages, Juntendo University, Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
| | - Kenta Izumi
- Department of Respiratory Medicine, Juntendo University, Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan; Research Institute for Diseases of Old Ages, Juntendo University, Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
| | - Koichiro Kanamori
- Department of Respiratory Medicine, Juntendo University, Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan; Research Institute for Diseases of Old Ages, Juntendo University, Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
| | - Yosuke Miyashita
- Department of Respiratory Medicine, Juntendo University, Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan; Research Institute for Diseases of Old Ages, Juntendo University, Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
| | - Takafumi Handa
- Department of Human Pathology, Juntendo University, Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
| | - Tetsuhiko Asao
- Department of Respiratory Medicine, Juntendo University, Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan; Research Institute for Diseases of Old Ages, Juntendo University, Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
| | - Ryo Ko
- Department of Respiratory Medicine, Juntendo University, Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan; Research Institute for Diseases of Old Ages, Juntendo University, Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
| | - Takehito Shukuya
- Department of Respiratory Medicine, Juntendo University, Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan; Research Institute for Diseases of Old Ages, Juntendo University, Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
| | - Naoko Shimada
- Department of Respiratory Medicine, Juntendo University, Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan; Research Institute for Diseases of Old Ages, Juntendo University, Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
| | - Kazuya Takamochi
- Department of General Thoracic Surgery, Juntendo University, Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
| | - Takuo Hayashi
- Department of Human Pathology, Juntendo University, Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
| | - Kenji Suzuki
- Department of General Thoracic Surgery, Juntendo University, Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
| | - Kazuhisa Takahashi
- Department of Respiratory Medicine, Juntendo University, Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan; Research Institute for Diseases of Old Ages, Juntendo University, Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
| |
Collapse
|
48
|
Murayama T, Mahadevan NR, Meador CB, Ivanova EV, Pan Y, Knelson EH, Tani T, Nakayama J, Ma X, Thai TC, Hung YP, Kim W, Watanabe H, Cai KQ, Hata AN, Paweletz CP, Barbie DA, Cañadas I. Targeting TREX1 Induces Innate Immune Response in Drug-Resistant Small-Cell Lung Cancer. CANCER RESEARCH COMMUNICATIONS 2024; 4:2399-2414. [PMID: 39177280 PMCID: PMC11391691 DOI: 10.1158/2767-9764.crc-24-0360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 07/23/2024] [Accepted: 08/21/2024] [Indexed: 08/24/2024]
Abstract
Small-cell lung cancer (SCLC) is the most lethal type of lung cancer. Paradoxically, this tumor displays an initial exquisite response to chemotherapy; however, at relapse, the tumor is highly resistant to subsequent available therapies. Here, we report that the expression of three prime repair exonuclease 1 (TREX1) is strongly induced in chemoresistant SCLCs. Assay for transposase-accessible chromatin using sequencing and chromatin immunoprecipitation sequencing revealed a significant increase in chromatin accessibility and transcriptional activity of TREX1 gene locus in chemoresistant SCLCs. Analyses of human SCLC tumors and patient-derived xenografts (PDX) also showed an increase in TREX1 expression in postchemotherapy samples. TREX1 depletion caused the activation of cyclic GMP-AMP synthase stimulator of interferon gene pathway due to cytoplasmic accumulation of damage-associated double-stranded DNA, inducing immunogenicity and enhancing the sensitivity of drug-resistant cells to chemotherapy. These findings suggest TREX1 upregulation may partially contribute to the survival of resistant cells, and its inhibition may represent a promising therapeutic strategy to enhance antitumor immunity and potentiate the efficacy of chemotherapy and/or immunotherapy in chemoresistant SCLCs. Significance: In this study, we show that targeting TREX1 induces an innate immune response and resensitizes SCLC cells to chemotherapy, representing a promising novel target for "immunologically" cold tumors, such as SCLC.
Collapse
Affiliation(s)
- Takahiko Murayama
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
- Cancer Epigenetics Institute, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Navin R Mahadevan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Catherine B Meador
- Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | - Elena V Ivanova
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Yuqiao Pan
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
- Cancer Epigenetics Institute, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Erik H Knelson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Tetsuo Tani
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Jun Nakayama
- Department of Oncogenesis and Growth Regulation, Osaka International Cancer Institute, Osaka, Japan
- Laboratory of Integrative Oncology, National Cancer Center Research Institute, Tokyo, Japan
| | - Xueying Ma
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
- Cancer Epigenetics Institute, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Tran C Thai
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Yin P Hung
- Department of Pathology, Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | - William Kim
- Moores Cancer Center, UC San Diego, La Jolla, California
- Center for Novel Therapeutics, UC San Diego, La Jolla, California
- Department of Medicine, UC San Diego, La Jolla, California
| | - Hideo Watanabe
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Kathy Q Cai
- Histopathology Facility, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Aaron N Hata
- Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | - Cloud P Paweletz
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - David A Barbie
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Israel Cañadas
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
- Cancer Epigenetics Institute, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| |
Collapse
|
49
|
Liu W, Li G, Huang D, Qin T. AKR1C3 promotes progression and mediates therapeutic resistance by inducing epithelial-mesenchymal transition and angiogenesis in small cell lung cancer. Transl Oncol 2024; 47:102027. [PMID: 38954974 PMCID: PMC11263718 DOI: 10.1016/j.tranon.2024.102027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/30/2024] [Accepted: 06/03/2024] [Indexed: 07/04/2024] Open
Abstract
OBJECTIVE Small cell lung cancer (SCLC) is a high-grade neuroendocrine tumor characterized by initial sensitivity to chemotherapy, followed by the development of drug resistance. The underlying mechanisms of resistance in SCLC have not been fully elucidated. Aldo-keto reductase family 1 member C3 (AKR1C3), is known to be associated with chemoradiotherapy resistance in diverse tumors. We aim to evaluate the prognostic significance and immune characteristics of AKR1C3 and investigate its potential role in promoting drug resistance in SCLC. METHODS 81 postoperative SCLC tissues were used to analyze AKR1C3 prognostic value and immune features. The tissue microarrays were employed to validate the clinical significance of AKR1C3 in SCLC. The effects of AKR1C3 on SCLC cell proliferation, migration, apoptosis and tumor angiogenesis were detected by CCK-8, wound healing assay, transwell assay, flow cytometry and tube formation assay. RESULTS AKR1C3 demonstrated the highest expression level compared to other AKR1C family genes, and multivariate cox regression analysis identified it as an independent prognostic factor for SCLC. High AKR1C3 expression patients who underwent chemoradiotherapy experienced significantly shorter overall survival (OS). Furthermore, AKR1C3 was involved in the regulation of the tumor immune microenvironment in SCLC. Silencing of AKR1C3 led to the inhibition of cell proliferation and migration, while simultaneously promoting apoptosis and reducing epithelial-mesenchymal transition (EMT) in SCLC. CONCLUSION AKR1C3 promotes cell growth and metastasis, leading to drug resistance through inducing EMT and angiogenesis in SCLC.
Collapse
Affiliation(s)
- Wenting Liu
- Department of Thoracic Oncology, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China; Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Guoli Li
- Department of Clinical Laboratory, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Dingzhi Huang
- Department of Thoracic Oncology, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China.
| | - Tingting Qin
- Department of Thoracic Oncology, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China.
| |
Collapse
|
50
|
Andrini E, Ricco G, Zappi A, Aloi S, Giordano M, Altimari A, Gruppioni E, Maloberti T, de Biase D, Campana D, Lamberti G. Challenges and future perspectives for the use of temozolomide in the treatment of SCLC. Cancer Treat Rev 2024; 129:102798. [PMID: 38970838 DOI: 10.1016/j.ctrv.2024.102798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 06/09/2024] [Accepted: 07/02/2024] [Indexed: 07/08/2024]
Abstract
Small-cell lung cancer (SCLC), accounting for 10-20 % of all lung tumors, represents the most aggressive high-grade neuroendocrine carcinoma. Most patients are diagnosed with extensive-stage SCLC (ES-SCLC), with brian metastases identified in ∼ 80 % of cases during the disease cours, and the prognosis is dismal, with a 5-year survival rate of less than 5 %. Current available treatments in the second-line setting are limited, and topotecan has long been the only FDA-approved drug in relapsed or refractory ES-SCLC, until the recent approval of lurbinectedin, a selective inhibitor of RNA polymerase II. Temozolomide (TMZ) is an oral alkylating agent, which showed single-agent activity in SCLC, particularly among patients with O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation. Several studies have revealed the synergistic activity of temozolomide with poly-ADP-ribose polymerase (PARP) inhibitors, that prevent repair of TMZ-induced DNA damage. This review focuses on the rationale for the use of TMZ in ES-SCLC and provides an overview of the main trials that have evaluated and are currently investigating its role, both as a single-agent and in combinations, in relapse or refractory disease.
Collapse
Affiliation(s)
- Elisa Andrini
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy.
| | - Gianluca Ricco
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy.
| | - Arianna Zappi
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy.
| | - Serena Aloi
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy.
| | - Mirela Giordano
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy.
| | - Annalisa Altimari
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.
| | - Elisa Gruppioni
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.
| | - Thais Maloberti
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.
| | - Dario de Biase
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy; Department of Pharmacy and Biotechnology (FaBit), University of Bologna, Bologna, Italy.
| | - Davide Campana
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy.
| | - Giuseppe Lamberti
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy.
| |
Collapse
|