251
|
Choi SH, Mani M, Kim J, Cho WJ, Martin TFJ, Kim JH, Chu HS, Jeong WJ, Won YW, Lee BJ, Ahn B, Kim J, Jeon DY, Park JW. DRG2 is required for surface localization of PD-L1 and the efficacy of anti-PD-1 therapy. Cell Death Discov 2024; 10:260. [PMID: 38802348 PMCID: PMC11130180 DOI: 10.1038/s41420-024-02027-x] [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: 01/25/2024] [Revised: 04/10/2024] [Accepted: 05/14/2024] [Indexed: 05/29/2024] Open
Abstract
More than half of tumor patients with high PD-L1 expression do not respond to anti-PD-1/PD-L1 therapy, and the underlying mechanisms are yet to be clarified. Here we show that developmentally regulated GTP-binding protein 2 (DRG2) is required for response of PD-L1-expressing tumors to anti-PD-1 therapy. DRG2 depletion enhanced IFN-γ signaling and increased the PD-L1 level in melanoma cells. However, it inhibited recycling of endosomal PD-L1 and reduced surface PD-L1 levels, which led to defects in interaction with PD-1. Anti-PD-1 did not expand effector-like T cells within DRG2-depleted tumors and failed to improve the survival of DRG2-depleted tumor-bearing mice. Cohort analysis revealed that patients bearing melanoma with low DRG2 protein levels were resistant to anti-PD-1 therapy. These findings identify DRG2 as a key regulator of recycling of endosomal PD-L1 and response to anti-PD-1 therapy and provide insights into how to increase the correlation between PD-L1 expression and response to anti-PD-1 therapy.
Collapse
Affiliation(s)
- Seong Hee Choi
- Department of Biological Sciences, University of Ulsan, Ulsan, Korea
- RopheLBio, B102, Seoul Forest M Tower, Seoul, Korea
| | - Muralidharan Mani
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Jeonghwan Kim
- School of System Biomedical Science, Soongsil University, Seoul, Korea
| | - Wha Ja Cho
- Department of Biological Sciences, University of Ulsan, Ulsan, Korea
| | - Thomas F J Martin
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Jee Hyun Kim
- RopheLBio, B102, Seoul Forest M Tower, Seoul, Korea
| | - Hun Su Chu
- RopheLBio, B102, Seoul Forest M Tower, Seoul, Korea
| | | | - Young-Wook Won
- RopheLBio, B102, Seoul Forest M Tower, Seoul, Korea
- Department of Biomedical Engineering, University of North Texas, Denton, TX, USA
| | - Byung Ju Lee
- Department of Biological Sciences, University of Ulsan, Ulsan, Korea
- Basic-Clinic Convergence Research Institute, University of Ulsan, Ulsan, Korea
| | - Byungyong Ahn
- Basic-Clinic Convergence Research Institute, University of Ulsan, Ulsan, Korea
- Department of Food Science and Nutrition, University of Ulsan, Ulsan, Korea
| | - Junil Kim
- School of System Biomedical Science, Soongsil University, Seoul, Korea.
| | - Do Yong Jeon
- Department of Biological Sciences, University of Ulsan, Ulsan, Korea.
| | - Jeong Woo Park
- Department of Biological Sciences, University of Ulsan, Ulsan, Korea.
- Basic-Clinic Convergence Research Institute, University of Ulsan, Ulsan, Korea.
| |
Collapse
|
252
|
Qiu J, Cheng Z, Jiang Z, Gan L, Zhang Z, Xie Z. Immunomodulatory Precision: A Narrative Review Exploring the Critical Role of Immune Checkpoint Inhibitors in Cancer Treatment. Int J Mol Sci 2024; 25:5490. [PMID: 38791528 PMCID: PMC11122264 DOI: 10.3390/ijms25105490] [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: 03/24/2024] [Revised: 05/11/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
An immune checkpoint is a signaling pathway that regulates the recognition of antigens by T-cell receptors (TCRs) during an immune response. These checkpoints play a pivotal role in suppressing excessive immune responses and maintaining immune homeostasis against viral or microbial infections. There are several FDA-approved immune checkpoint inhibitors (ICIs), including ipilimumab, pembrolizumab, and avelumab. These ICIs target cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), programmed cell death protein 1 (PD-1), and programmed death ligand 1 (PD-L1). Furthermore, ongoing efforts are focused on developing new ICIs with emerging potential. In comparison to conventional treatments, ICIs offer the advantages of reduced side effects and durable responses. There is growing interest in the potential of combining different ICIs with chemotherapy, radiation therapy, or targeted therapies. This article comprehensively reviews the classification, mechanism of action, application, and combination strategies of ICIs in various cancers and discusses their current limitations. Our objective is to contribute to the future development of more effective anticancer drugs targeting immune checkpoints.
Collapse
Affiliation(s)
- Junyu Qiu
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (J.Q.); (Z.C.); (Z.J.); (L.G.); (Z.Z.)
- Queen Mary School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Zilin Cheng
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (J.Q.); (Z.C.); (Z.J.); (L.G.); (Z.Z.)
- Queen Mary School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Zheng Jiang
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (J.Q.); (Z.C.); (Z.J.); (L.G.); (Z.Z.)
- Queen Mary School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Luhan Gan
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (J.Q.); (Z.C.); (Z.J.); (L.G.); (Z.Z.)
- Huan Kui School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Zixuan Zhang
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (J.Q.); (Z.C.); (Z.J.); (L.G.); (Z.Z.)
- Queen Mary School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Zhenzhen Xie
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (J.Q.); (Z.C.); (Z.J.); (L.G.); (Z.Z.)
| |
Collapse
|
253
|
Lee E, Jang JY, Yang J. Uncommon Adverse Events of Immune Checkpoint Inhibitors in Small Cell Lung Cancer: A Systematic Review of Case Reports. Cancers (Basel) 2024; 16:1896. [PMID: 38791974 PMCID: PMC11119772 DOI: 10.3390/cancers16101896] [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: 04/08/2024] [Revised: 05/03/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
BACKGROUND This study aimed to systematically review case reports documenting rare adverse events in patients with small cell lung cancer (SCLC) following the administration of immune checkpoint inhibitors (ICIs). METHODS A systematic literature review was conducted to identify case reports detailing previously unreported adverse drug reactions to ICIs in patients with SCLC. The scope of the literature reviewed was restricted to case studies on SCLC published up to 31 December 2023. RESULTS We analyzed twenty-four studies on ICI use for patients with SCLC. There were six reports on atezolizumab, four on durvalumab, and three on adverse events from monotherapy with nivolumab. Reports involving combination treatments were the most frequent, with a total of six, predominantly involving using nivolumab in combination with ipilimumab. Additionally, there was one report each on using pembrolizumab, nofazinilimab, sintilimab, tislelizumab, and toripalimab. We collected detailed information on the clinical course, including patient and disease characteristics, symptoms, treatment for each adverse event, and recovery status. Among the patients included in the case reports, 21 out of 24 (87.5%) had extensive-stage SCLC when initiating ICI therapy, with only 1 patient diagnosed with limited-stage SCLC. Respiratory system adverse events were most common, with seven cases, followed by neurological, endocrinological, and gastroenterological events. Three case reports documented adverse events across multiple systems in a single patient. In most cases, patients showed symptom improvement; however, four studies reported cases where patients either expired without symptom improvement or experienced sequelae. CONCLUSIONS Efforts to develop reliable biomarkers for predicting irAEs continue, with ongoing research to enhance predictive precision. Immunotherapy presents diverse and unpredictable adverse events, underscoring the need for advanced diagnostic tools and a multidisciplinary approach to improve patient management.
Collapse
Affiliation(s)
- Eunso Lee
- Division of Allergy and Pulmonology, Department of Internal Medicine, Chungnam National University Sejong Hospital, Sejong 30099, Republic of Korea
| | - Jeong Yun Jang
- Department of Radiation Oncology, Konkuk University Medical Center, Konkuk University School of Medicine, 120-1, Neungdong-ro, Gwangjin-gu, Seoul 05030, Republic of Korea
| | - Jinho Yang
- Department of Occupational Health and Safety, Semyung University, 65 Semyung-ro, Jecheon 27136, Republic of Korea
| |
Collapse
|
254
|
Hosseini I, Fleisher B, Getz J, Decalf J, Kwong M, Ovacik M, Bainbridge TW, Moussion C, Rao GK, Gadkar K, Kamath AV, Ramanujan S. A Minimal PBPK/PD Model with Expansion-Enhanced Target-Mediated Drug Disposition to Support a First-in-Human Clinical Study Design for a FLT3L-Fc Molecule. Pharmaceutics 2024; 16:660. [PMID: 38794321 PMCID: PMC11125320 DOI: 10.3390/pharmaceutics16050660] [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: 03/28/2024] [Revised: 04/27/2024] [Accepted: 05/03/2024] [Indexed: 05/26/2024] Open
Abstract
FLT3L-Fc is a half-life extended, effectorless Fc-fusion of the native human FLT3-ligand. In cynomolgus monkeys, treatment with FLT3L-Fc leads to a complex pharmacokinetic/pharmacodynamic (PK/PD) relationship, with observed nonlinear PK and expansion of different immune cell types across different dose levels. A minimal physiologically based PK/PD model with expansion-enhanced target-mediated drug disposition (TMDD) was developed to integrate the molecule's mechanism of action, as well as the complex preclinical and clinical PK/PD data, to support the preclinical-to-clinical translation of FLT3L-Fc. In addition to the preclinical PK data of FLT3L-Fc in cynomolgus monkeys, clinical PK and PD data from other FLT3-agonist molecules (GS-3583 and CDX-301) were used to inform the model and project the expansion profiles of conventional DC1s (cDC1s) and total DCs in peripheral blood. This work constitutes an essential part of our model-informed drug development (MIDD) strategy for clinical development of FLT3L-Fc by projecting PK/PD in healthy volunteers, determining the first-in-human (FIH) dose, and informing the efficacious dose in clinical settings. Model-generated results were incorporated in regulatory filings to support the rationale for the FIH dose selection.
Collapse
|
255
|
Shalata W, Maimon Rabinovich N, Agbarya A, Yakobson A, Dudnik Y, Abu Jama A, Cohen AY, Shalata S, Abu Hamed A, Ilan Ber T, Machluf O, Shoham Levin G, Meirovitz A. Efficacy of Pembrolizumab vs. Nivolumab Plus Ipilimumab in Metastatic NSCLC in Relation to PD-L1 and TMB Status. Cancers (Basel) 2024; 16:1825. [PMID: 38791905 PMCID: PMC11119071 DOI: 10.3390/cancers16101825] [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: 04/08/2024] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
The efficacy of immune checkpoint inhibitor (ICI) therapy concerning programmed death ligand 1 (PD-L1) status is well established in patients diagnosed with non-small cell lung cancer (NSCLC). However, there remains a paucity of evidence regarding the efficacy concerning tumor mutational burden (TMB) in both clinical trials and real-world data (RWD). In the current article, clinicopathological and molecular epidemiological data were meticulously collected, and treatment modalities were meticulously recorded. The final analysis included a study population of 194 patients. Median age was 67 years (range 37-86), with the majority being male (71.13%), and 85.71% of patients were either current or former smokers at diagnosis. Adenocarcinoma accounted for most diagnoses (71.65%), followed by squamous cell carcinoma (24.23%). In terms of PD-L1 status, 42.78% had an expression level below 1%, 28.35% had an expression between 1-49%, and 28.87% had an expression above 50%. The TMB ranged from 0 to 75, with a median of 10.31 (range 0-75) for PD-L1 expression below 1%, with a median of 9.73 (range 0.95-39.63) for PD-L1 expression between 1-49%, and a median of 9.72 (range 0.95-48) for PD-L1 expression above 50%. Corresponding to patients with low PDL-1 less than 1% and low TMB (0-5), the median overall survival (mOS) was 16 (p = 0.18), and 15 months (p = 0.22), patients with medium PDL-1 (1-49%) and medium TMB (5-10), the mOS was 15 (p = 0.18) and 16 months (p = 0.22), patients with high PDL-1 (>50) and high TMB (>10), the mOS was 24 (p = 0.18) and 21 (p = 0.22) months. This study represents the largest academic RWD dataset concerning PD-L1 and TMB status in patients with locally advanced and metastatic NSCLC.
Collapse
Affiliation(s)
- Walid Shalata
- The Legacy Heritage Cancer Center and Dr. Larry Norton Institute, Soroka Medical Center, Beer-Sheva 84105, Israel
- Medical School for International Health, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | | | - Abed Agbarya
- Oncology Department, Bnai Zion Medical Center, Haifa 31048, Israel
| | - Alexander Yakobson
- The Legacy Heritage Cancer Center and Dr. Larry Norton Institute, Soroka Medical Center, Beer-Sheva 84105, Israel
- Medical School for International Health, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Yulia Dudnik
- The Legacy Heritage Cancer Center and Dr. Larry Norton Institute, Soroka Medical Center, Beer-Sheva 84105, Israel
- Medical School for International Health, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Ashraf Abu Jama
- The Legacy Heritage Cancer Center and Dr. Larry Norton Institute, Soroka Medical Center, Beer-Sheva 84105, Israel
- Medical School for International Health, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Ahron Yehonatan Cohen
- The Legacy Heritage Cancer Center and Dr. Larry Norton Institute, Soroka Medical Center, Beer-Sheva 84105, Israel
- Medical School for International Health, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Sondos Shalata
- Nutrition Unit, Galilee Medical Center, Nahariya 22000, Israel;
| | - Ahmad Abu Hamed
- The Legacy Heritage Cancer Center and Dr. Larry Norton Institute, Soroka Medical Center, Beer-Sheva 84105, Israel
| | | | | | | | - Amichay Meirovitz
- The Legacy Heritage Cancer Center and Dr. Larry Norton Institute, Soroka Medical Center, Beer-Sheva 84105, Israel
- Medical School for International Health, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| |
Collapse
|
256
|
Hou J, Xie S, Gao J, Jiang T, Zhu E, Yang X, Jin Z, Long H, Zhang A, Yang F, Wang L, Zha H, Jia Q, Zhu B, Wang X. NK cell transfer overcomes resistance to PD-(L)1 therapy in aged mice. Exp Hematol Oncol 2024; 13:48. [PMID: 38725070 PMCID: PMC11080179 DOI: 10.1186/s40164-024-00511-9] [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: 12/19/2023] [Accepted: 04/08/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Cancer is the leading cause of death among older adults. Although the integration of immunotherapy has revolutionized the therapeutic landscape of cancer, the complex interactions between age and immunotherapy efficacy remain incompletely defined. Here, we aimed to elucidate the relationship between aging and immunotherapy resistance. METHODS Flow cytometry was performed to evaluate the infiltration of immune cells in the tumor microenvironment (TME). In vivo T cell proliferation, cytotoxicity and migration assays were performed to evaluate the antitumor capacity of tumor antigen-specific CD8+ T cells in mice. Real-time quantitative PCR (qPCR) was used to investigate the expression of IFN-γ-associated gene and natural killer (NK)-associated chemokine. Adoptive NK cell transfer was adopted to evaluate the effects of NK cells from young mice in overcoming the immunotherapy resistance of aged mice. RESULTS We found that elderly patients with advanced non-small cell lung cancer (aNSCLC) aged ≥ 75 years exhibited poorer progression-free survival (PFS), overall survival (OS) and a lower clinical response rate after immunotherapy. Mechanistically, we showed that the infiltration of NK cells was significantly reduced in aged mice compared to younger mice. Furthermore, the aged NK cells could also suppress the activation of tumor antigen-specific CD8+ T cells by inhibiting the recruitment and activation of CD103+ dendritic cells (DCs). Adoptive transfer of NK cells from young mice to aged mice promoted TME remodeling, and reversed immunotherapy resistance. CONCLUSION Our findings revealed the decreased sensitivity of elderly patients to immunotherapy, as well as in aged mice. This may be attributed to the reduction of NK cells in aged mice, which inhibits CD103+ DCs recruitment and its CD86 expression and ultimately leads to immunotherapy resistance.
Collapse
Affiliation(s)
- Junlei Hou
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Shuanglong Xie
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
- Jinfeng Laboratory, Chongqing, 401329, China
| | - Jianbao Gao
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Tao Jiang
- Shanghai Pulmonary Hospital, Shanghai, 200082, China
| | - Enjian Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Xuezhi Yang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Zheng Jin
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Haixia Long
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Anmei Zhang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Fei Yang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Lujing Wang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Haoran Zha
- Department of Oncology, PLA Rocket Force Characteristic Medical Center, Beijing, 100088, China
| | - Qingzhu Jia
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Xinxin Wang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| |
Collapse
|
257
|
Zhao P, Zhao T, Yu L, Ma W, Liu W, Zhang C. The risk of endocrine immune-related adverse events induced by PD-1 inhibitors in cancer patients: a systematic review and meta-analysis. Front Oncol 2024; 14:1381250. [PMID: 38756658 PMCID: PMC11096456 DOI: 10.3389/fonc.2024.1381250] [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: 02/03/2024] [Accepted: 04/17/2024] [Indexed: 05/18/2024] Open
Abstract
Objective Endocrinopathies are the most common immune-related adverse events (irAEs) observed during therapy with PD-1 inhibitors. In this study, we conducted a comprehensive systematic review and meta-analysis to evaluate the risk of immune-related endocrinopathies in patients treated with PD-1 inhibitors. Methods We performed a systematic search in the PubMed, Embase, and Cochrane Library databases to retrieve all randomized controlled trials (RCTs) involving PD-1 inhibitors, spanning from their inception to November 24, 2023. The comparative analysis encompassed patients undergoing chemotherapy, targeted therapy, or receiving placebo as control treatments. This study protocol has been registered with PROSPERO (CRD42023488303). Results A total of 48 clinical trials comprising 24,514 patients were included. Compared with control groups, patients treated with PD-1 inhibitors showed an increased risk of immune-related adverse events, including hypothyroidism, hyperthyroidism, hypophysitis, thyroiditis, diabetes mellitus, and adrenal insufficiency. Pembrolizumab was associated with an increased risk of all aforementioned endocrinopathies (hypothyroidism: RR=4.76, 95%CI: 3.55-6.39; hyperthyroidism: RR=9.69, 95%CI: 6.95-13.52; hypophysitis: RR=5.47, 95%CI: 2.73-10.97; thyroiditis: RR=5.95, 95%CI: 3.02-11.72; diabetes mellitus: RR=3.60, 95%CI: 1.65-7.88; adrenal insufficiency: RR=4.80, 95%CI: 2.60-8.88). Nivolumab was associated with an increased risk of hypothyroidism (RR=7.67, 95%CI: 5.00-11.75) and hyperthyroidism (RR=9.22, 95%CI: 4.71-18.04). Tislelizumab and sintilimab were associated with an increased risk of hypothyroidism (RR=19.07, 95%CI: 5.46-66.69 for tislelizumab and RR=18.36, 95%CI: 3.58-94.21 for sintilimab). For different tumor types, both hypothyroidism and hyperthyroidism were at high risks. Besides, patients with non-small cell lung cancer were at a higher risk of thyroiditis and adrenal insufficiency. Patients with melanoma were at a higher risk of hypophysitis and diabetes mellitus. Both low- and high-dose group increased risks of hypothyroidism and hyperthyroidism. Conclusion Risk of endocrine irAEs may vary in different PD-1 inhibitors and different tumor types. Increased awareness and understanding of the risk features of endocrine irAEs associated with PD-1 inhibitors is critical for clinicians. Systematic review registration crd.york.ac.uk/prospero, identifier PROSPERO (CRD42023488303).
Collapse
Affiliation(s)
- Pengfei Zhao
- Department of Clinical Pharmacy, Weifang People's Hospital, Weifang, China
| | - Ting Zhao
- Department of Clinical Pharmacy, Weifang People's Hospital, Weifang, China
| | - Lihong Yu
- Department of Clinical Pharmacy, Weifang People's Hospital, Weifang, China
| | - Wenming Ma
- Department of Clinical Pharmacy, Weifang People's Hospital, Weifang, China
| | - Wenyu Liu
- Department of Pharmacy, Weifang People's Hospital, Weifang, China
| | - Chenning Zhang
- Department of Rehabilitation Medicine & Department of Pharmacy, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, China
| |
Collapse
|
258
|
Tangudu NK, Buj R, Wang H, Wang J, Cole AR, Uboveja A, Fang R, Amalric A, Yang B, Chatoff A, Crispim CV, Sajjakulnukit P, Lyons MA, Cooper K, Hempel N, Lyssiotis CA, Chandran UR, Snyder NW, Aird KM. De Novo Purine Metabolism is a Metabolic Vulnerability of Cancers with Low p16 Expression. CANCER RESEARCH COMMUNICATIONS 2024; 4:1174-1188. [PMID: 38626341 PMCID: PMC11064835 DOI: 10.1158/2767-9764.crc-23-0450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 03/04/2024] [Accepted: 04/11/2024] [Indexed: 04/18/2024]
Abstract
p16 is a tumor suppressor encoded by the CDKN2A gene whose expression is lost in approximately 50% of all human cancers. In its canonical role, p16 inhibits the G1-S-phase cell cycle progression through suppression of cyclin-dependent kinases. Interestingly, p16 also has roles in metabolic reprogramming, and we previously published that loss of p16 promotes nucleotide synthesis via the pentose phosphate pathway. However, the broader impact of p16/CDKN2A loss on other nucleotide metabolic pathways and potential therapeutic targets remains unexplored. Using CRISPR knockout libraries in isogenic human and mouse melanoma cell lines, we determined several nucleotide metabolism genes essential for the survival of cells with loss of p16/CDKN2A. Consistently, many of these genes are upregulated in melanoma cells with p16 knockdown or endogenously low CDKN2A expression. We determined that cells with low p16/CDKN2A expression are sensitive to multiple inhibitors of de novo purine synthesis, including antifolates. Finally, tumors with p16 knockdown were more sensitive to the antifolate methotrexate in vivo than control tumors. Together, our data provide evidence to reevaluate the utility of these drugs in patients with p16/CDKN2Alow tumors as loss of p16/CDKN2A may provide a therapeutic window for these agents. SIGNIFICANCE Antimetabolites were the first chemotherapies, yet many have failed in the clinic due to toxicity and poor patient selection. Our data suggest that p16 loss provides a therapeutic window to kill cancer cells with widely-used antifolates with relatively little toxicity.
Collapse
Affiliation(s)
- Naveen Kumar Tangudu
- Department of Pharmacology and Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Raquel Buj
- Department of Pharmacology and Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Hui Wang
- Department of Pharmacology and Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Jiefei Wang
- Department of Biomedical Informatics and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Aidan R. Cole
- Department of Pharmacology and Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Apoorva Uboveja
- Department of Pharmacology and Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Richard Fang
- Department of Pharmacology and Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Amandine Amalric
- Department of Pharmacology and Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Baixue Yang
- Department of Pharmacology and Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Tsinghua University School of Medicine, Beijing, P.R. China
| | - Adam Chatoff
- Department of Cardiovascular Sciences, Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Claudia V. Crispim
- Department of Cardiovascular Sciences, Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Peter Sajjakulnukit
- Department of Molecular and Integrative Physiology, Department of Internal Medicine, Division of Gastroenterology, and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Maureen A. Lyons
- Genomics Facility, UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Kristine Cooper
- Biostatistics Facility, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Nadine Hempel
- Division of Hematology/Oncology, Department of Medicine, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Costas A. Lyssiotis
- Department of Molecular and Integrative Physiology, Department of Internal Medicine, Division of Gastroenterology, and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Uma R. Chandran
- Department of Biomedical Informatics and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Nathaniel W. Snyder
- Department of Cardiovascular Sciences, Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Katherine M. Aird
- Department of Pharmacology and Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
259
|
Reger De Moura C, Louveau B, Jouenne F, Vilquin P, Battistella M, Bellahsen-Harrar Y, Sadoux A, Menashi S, Dumaz N, Lebbé C, Mourah S. Inactivation of kindlin-3 increases human melanoma aggressiveness through the collagen-activated tyrosine kinase receptor DDR1. Oncogene 2024; 43:1620-1630. [PMID: 38570692 DOI: 10.1038/s41388-024-03014-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 03/15/2024] [Accepted: 03/21/2024] [Indexed: 04/05/2024]
Abstract
The role of the focal adhesion protein kindlin-3 as a tumor suppressor and its interaction mechanisms with extracellular matrix constitute a major field of investigation to better decipher tumor progression. Besides the well-described role of kindlin-3 in integrin activation, evidence regarding modulatory functions between melanoma cells and tumor microenvironment are lacking and data are needed to understand mechanisms driven by kindlin-3 inactivation. Here, we show that kindlin-3 inactivation through knockdown or somatic mutations increases BRAFV600mut melanoma cells oncogenic properties via collagen-related signaling by decreasing cell adhesion and enhancing proliferation and migration in vitro, and by promoting tumor growth in mice. Mechanistic analysis reveals that kindlin-3 interacts with the collagen-activated tyrosine kinase receptor DDR1 (Discoidin domain receptor 1) modulating its expression and its interaction with β1-integrin. Kindlin-3 knockdown or mutational inactivation disrupt DDR1/β1-integrin complex in vitro and in vivo and its loss improves the anti-proliferative effect of DDR1 inhibition. In agreement, kindlin-3 downregulation is associated with DDR1 over-expression in situ and linked to worse melanoma prognosis. Our study reveals a unique mechanism of action of kindlin-3 in the regulation of tumorigenesis mediated by the collagen-activated tyrosine kinase receptor DDR1 thus paving the way for innovative therapeutic targeting approaches in melanoma.
Collapse
Affiliation(s)
- Coralie Reger De Moura
- Department of Pharmacology and Tumor Genomics, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France
- Université Paris Cité, INSERM UMR-S 976, Team 1, Human Immunology Pathophysiology & Immunotherapy (HIPI), F-75010, Paris, France
| | - Baptiste Louveau
- Department of Pharmacology and Tumor Genomics, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France
- Université Paris Cité, INSERM UMR-S 976, Team 1, Human Immunology Pathophysiology & Immunotherapy (HIPI), F-75010, Paris, France
| | - Fanélie Jouenne
- Department of Pharmacology and Tumor Genomics, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France
- Université Paris Cité, INSERM UMR-S 976, Team 1, Human Immunology Pathophysiology & Immunotherapy (HIPI), F-75010, Paris, France
| | - Paul Vilquin
- Department of Pharmacology and Tumor Genomics, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France
| | - Maxime Battistella
- Department of Pathology, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France
| | - Yaelle Bellahsen-Harrar
- Department of Pathology, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France
| | - Aurélie Sadoux
- Department of Pharmacology and Tumor Genomics, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France
| | - Suzanne Menashi
- Department of Pharmacology and Tumor Genomics, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France
- Université Paris Cité, INSERM UMR-S 976, Team 1, Human Immunology Pathophysiology & Immunotherapy (HIPI), F-75010, Paris, France
| | - Nicolas Dumaz
- Université Paris Cité, INSERM UMR-S 976, Team 1, Human Immunology Pathophysiology & Immunotherapy (HIPI), F-75010, Paris, France
| | - Céleste Lebbé
- Université Paris Cité, INSERM UMR-S 976, Team 1, Human Immunology Pathophysiology & Immunotherapy (HIPI), F-75010, Paris, France
- Department of Dermatology and CIC, Hôpital Saint Louis, Cancer Institute, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France
| | - Samia Mourah
- Department of Pharmacology and Tumor Genomics, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France.
- Université Paris Cité, INSERM UMR-S 976, Team 1, Human Immunology Pathophysiology & Immunotherapy (HIPI), F-75010, Paris, France.
| |
Collapse
|
260
|
Albrecht LJ, Dimitriou F, Grover P, Hassel JC, Erdmann M, Forschner A, Johnson DB, Váraljai R, Lodde G, Placke JM, Krefting F, Zaremba A, Ugurel S, Roesch A, Schulz C, Berking C, Pöttgen C, Menzies AM, Long GV, Dummer R, Livingstone E, Schadendorf D, Zimmer L. Anti-PD-(L)1 plus BRAF/MEK inhibitors (triplet therapy) after failure of immune checkpoint inhibition and targeted therapy in patients with advanced melanoma. Eur J Cancer 2024; 202:113976. [PMID: 38484692 DOI: 10.1016/j.ejca.2024.113976] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 02/16/2024] [Accepted: 02/21/2024] [Indexed: 04/21/2024]
Abstract
BACKGROUND Effective treatment options are limited for patients with advanced melanoma who have progressed on immune checkpoint inhibitors (ICI) and targeted therapies (TT). Preclinical models support the combination of ICI with TT; however, clinical trials evaluating the efficacy of triplet combinations in first-line setting showed limited advantage compared to TT only. METHODS We conducted a retrospective, multicenter study, that included patients with advanced melanoma who were treated with BRAF/MEK inhibitors in combination with an anti-PD-(L)1 antibody (triplet therapy) after failure of at least one anti-PD-(L)1-based therapy and one TT in seven major melanoma centers between February 2016 and July 2022. RESULTS A total of 48 patients were included, of which 32 patients, 66.7% had brain metastases, 37 patients (77.1%) had three or more metastatic organs and 21 patients (43.8%) had three or more treatment lines. The median follow-up time was 31.4 months (IQR, 22.27-40.45 months). The treatment with triplet therapy resulted in an ORR of 35.4% (n = 17) and a DCR of 47.9% (n = 23). The median DOR was 5.9 months (range, 3.39-14.27 months). Patients treated with BRAF/MEK inhibitors as the last treatment line showed a slightly lower ORR (29.6%) compared to patients who received ICI or chemotherapy last (ORR: 42.9%). Grade 3-4 treatment-related adverse events occurred in 25% of patients (n = 12), with seven patients (14.6%) requiring discontinuation of treatment with both or either drug. CONCLUSIONS Triplet therapy has shown activity in heavily pretreated patients with advanced melanoma and may represent a potential treatment regimen after failure of ICI and TT.
Collapse
Affiliation(s)
- Lea Jessica Albrecht
- Department of Dermatology, University Hospital Essen, West German Cancer Center, University Duisburg-Essen and the German Cancer Consortium (DKTK), Essen, Germany
| | - Florentia Dimitriou
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland
| | - Piyush Grover
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
| | - Jessica C Hassel
- Department of Dermatology and National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| | - Michael Erdmann
- Department of Dermatology, Uniklinikum Erlangen and the Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany
| | - Andrea Forschner
- Department of Dermatology, University Hospital Tuebingen, Tuebingen, Germany
| | - Douglas B Johnson
- Department of Medicine, Division of Hematology and Oncology, VUMC, and Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Renáta Váraljai
- Department of Dermatology, University Hospital Essen, West German Cancer Center, University Duisburg-Essen and the German Cancer Consortium (DKTK), Essen, Germany
| | - Georg Lodde
- Department of Dermatology, University Hospital Essen, West German Cancer Center, University Duisburg-Essen and the German Cancer Consortium (DKTK), Essen, Germany
| | - Jan Malte Placke
- Department of Dermatology, University Hospital Essen, West German Cancer Center, University Duisburg-Essen and the German Cancer Consortium (DKTK), Essen, Germany
| | - Frederik Krefting
- Department of Dermatology, University Hospital Essen, West German Cancer Center, University Duisburg-Essen and the German Cancer Consortium (DKTK), Essen, Germany
| | - Anne Zaremba
- Department of Dermatology, University Hospital Essen, West German Cancer Center, University Duisburg-Essen and the German Cancer Consortium (DKTK), Essen, Germany
| | - Selma Ugurel
- Department of Dermatology, University Hospital Essen, West German Cancer Center, University Duisburg-Essen and the German Cancer Consortium (DKTK), Essen, Germany
| | - Alexander Roesch
- Department of Dermatology, University Hospital Essen, West German Cancer Center, University Duisburg-Essen and the German Cancer Consortium (DKTK), Essen, Germany
| | - Carsten Schulz
- Department of Dermatology and National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| | - Carola Berking
- Department of Dermatology, Uniklinikum Erlangen and the Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany
| | - Christoph Pöttgen
- Department of Radiotherapy, West German Cancer Centre, University Hospital Essen, Essen, Germany
| | - Alexander M Menzies
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia; Department of Medical Oncology, Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia; Department of Medical Oncology, Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia
| | - Reinhard Dummer
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland
| | - Elisabeth Livingstone
- Department of Dermatology, University Hospital Essen, West German Cancer Center, University Duisburg-Essen and the German Cancer Consortium (DKTK), Essen, Germany
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, West German Cancer Center, University Duisburg-Essen and the German Cancer Consortium (DKTK), Essen, Germany; National Center for Tumor Diseases (NCT)-West, Campus Essen, & Research Alliance Ruhr, Research Center One Health, University Duisburg-Essen, Essen, Germany
| | - Lisa Zimmer
- Department of Dermatology, University Hospital Essen, West German Cancer Center, University Duisburg-Essen and the German Cancer Consortium (DKTK), Essen, Germany.
| |
Collapse
|
261
|
Jeung YS, Chun JY, Choi BK, Park SY, Lim HJ, Park JW, Han JY, Lee Y. Infection-related Hospitalizations During Immune Checkpoint Inhibitor Treatment Without Immunosuppressants. J Immunother 2024; 47:139-147. [PMID: 38282479 DOI: 10.1097/cji.0000000000000504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 11/13/2023] [Indexed: 01/30/2024]
Abstract
Immunosuppressants are increasingly being used in the clinic to manage immune-related adverse effects. Consequently, the incidence of secondary infections associated with immunosuppression is increasing. However, little is known about primary infections during immune checkpoint inhibitor (ICI) treatment without immunosuppressants. We aimed to evaluate primary infectious diseases during antiprogrammed death ligand-1 immunotherapy without immunosuppressants. We retrospectively screened medical records of 233 patients who underwent ICI treatment for advanced non-small cell lung cancer between January 2014 and May 2018 at National Cancer Center, Republic of Korea. Subsequently, we evaluated the clinical characteristics and treatment outcomes of selected patients hospitalized for potential infectious disease without immunosuppressive treatment (n=80). Eight cases (3.4%) were identified as bacterial pneumonia (n=5) and cellulitis, inflamed epidermoid cyst, and wound infection (n=1 each). The bacterial pathogens Streptococcus pneumoniae and Haemophilus influenzae were identified in 4 patients with pneumonia. The period between the start of ICI treatment and infection varied between 3 and 189 days (median, 24.5 days). Five (62.5%) patients were infected within a month after ICI treatment initiation. All patients were treated with empirical antibiotics and discharged without complications. The median progression-free and overall survival for ICI treatment was 11.5 and 25.5 months, respectively. Six patients experienced ICI-associated adverse effects postinfection: Herpes zoster infection (n=4) and pneumonitis (n=2). Infectious disease independent of immunosuppression is a rare, but possible event in patients with lung cancer receiving ICI treatment. Clinical awareness would enable prompt diagnosis of primary infection during immunotherapy.
Collapse
Affiliation(s)
- Ye Sul Jeung
- Department of Internal Medicine, National Cancer Center, Goyang, Republic of Korea
| | - June Young Chun
- Department of Internal Medicine, National Cancer Center, Goyang, Republic of Korea
| | - Beom Kyu Choi
- Biomedicine Production Branch, National Cancer Center, Goyang, Republic of Korea
| | - Seog Yun Park
- Department of Pathology, National Cancer Center, Goyang, Republic of Korea
| | - Hyun-Ju Lim
- Department of Diagnostic Radiology, National Cancer Center, Goyang, Republic of Korea
| | - Jong Woong Park
- Department of Orthopaedic Surgery, National Cancer Center, Goyang, Republic of Korea
| | - Ji-Youn Han
- Department of Internal Medicine, National Cancer Center, Goyang, Republic of Korea
| | - Youngjoo Lee
- Department of Internal Medicine, National Cancer Center, Goyang, Republic of Korea
| |
Collapse
|
262
|
Rydén V, El-Naggar AI, Koliadi A, Ladjevardi CO, Digkas E, Valachis A, Ullenhag GJ. The role of dacarbazine and temozolomide therapy after treatment with immune checkpoint inhibitors in malignant melanoma patients: A case series and meta-analysis. Pigment Cell Melanoma Res 2024; 37:352-362. [PMID: 38158376 DOI: 10.1111/pcmr.13156] [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: 09/13/2023] [Revised: 11/17/2023] [Accepted: 12/12/2023] [Indexed: 01/03/2024]
Abstract
Dacarbazine (DTIC) and its oral counterpart temozolomide (TMZ) have been the most used agents in advanced malignant melanoma (MM) patients and they are still used routinely. The preferred first line treatment, immune checkpoint inhibitors (CPIs) might shape the tumor and the tumor microenvironment, possibly affecting the response to subsequent therapies. The aim of this study was to investigate the treatment effect of DTIC/TMZ in MM patients after CPI therapy in a consecutive patient cohort and through systematic literature review and meta-analysis. Thirty-five patients with advanced MM treated with DTIC/TMZ after previous CPI therapy in three Swedish regions between 2017 and 2021 were recognized and seven case series studies were identified through systematic database review. Pooled data from all 345 patients showed a median real-world progression-free survival (rwPFS) of 1.9 months and overall survival (OS) of 6.0 months. Three of these studies were included in a meta-analysis comparing DTIC/TMZ after CPI treatment, versus no previous immunotherapy, showing no statistically significant differences in rwPFS or OS but higher real-world response rate to chemotherapy for the prior-CPI treated group (Odds Ratio: 2.24; 95% Confidence Interval: 1.04-4.86). The current study supports consideration of DTIC/TMZ in later line of treatment in the immunotherapy era.
Collapse
Affiliation(s)
- Viktoria Rydén
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Department of Oncology, Uppsala University Hospital, Uppsala, Sweden
| | - Ali Inan El-Naggar
- Department of Oncology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- Department of Oncology, Örebro University Hospital, Örebro, Sweden
| | - Anthoula Koliadi
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Department of Oncology, Uppsala University Hospital, Uppsala, Sweden
| | - Cecilia Olsson Ladjevardi
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Department of Oncology, Uppsala University Hospital, Uppsala, Sweden
| | - Evangelos Digkas
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Department of Oncology, Uppsala University Hospital, Uppsala, Sweden
- Department of Oncology, Eskilstuna, Sweden
| | - Antonios Valachis
- Department of Oncology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- Department of Oncology, Örebro University Hospital, Örebro, Sweden
| | - Gustav J Ullenhag
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Department of Oncology, Uppsala University Hospital, Uppsala, Sweden
| |
Collapse
|
263
|
Leeneman B, Xander NSH, Fiets WE, de Jong WK, Uyl NEM, Wymenga ANM, Reyners AKL, Uyl-de Groot CA. Assessing the clinical benefit of systemic anti-cancer treatments in the Netherlands: The impact of different thresholds for effectiveness. Eur J Cancer 2024; 202:114002. [PMID: 38489860 DOI: 10.1016/j.ejca.2024.114002] [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: 01/16/2024] [Revised: 02/27/2024] [Accepted: 02/28/2024] [Indexed: 03/17/2024]
Abstract
BACKGROUND In the Netherlands, the clinical benefit of systemic anti-cancer treatments (SACTs) is assessed by the Committee for the Evaluation of Oncological Agents (cieBOM). For non-curative SACTs, the assessment is based on the hazard ratio (HR) for progression-free survival and/or overall survival (OS), and the difference in median survival. We evaluated the impact of different thresholds for effectiveness by reassessing the clinical benefit of SACTs. METHODS We reassessed SACTs that were initially assessed by cieBOM between 2015 and 2017. Four scenarios were formulated: replacing an "OR" approach (initial assessment) by an "AND" approach (used in all scenarios), changing the HR threshold from < 0.70 (initial assessment) to < 0.60, changing the threshold for the difference in median survival from > 12 weeks (initial assessment) to > 16 weeks, and including thresholds for OS rates. The outcomes of these scenarios were compared to the outcomes of the initial assessment. RESULTS Reassessments were conducted for 41 treatments. Replacing the "OR" approach by an "AND" approach substantially decreased the number of positive assessments (from 33 to 22), predominantly affecting immunotherapies. This number further decreased (to 21 and 19, respectively) in case more restrictive thresholds for the HR and difference in median survival were used. Including thresholds for OS rates slightly mitigated the impact of applying an "AND" approach. CONCLUSIONS The scenario-specific thresholds had a substantial impact; the number of negative assessments more than doubled. Since this was not limited to treatments with marginal survival benefits, understanding the potential challenges that may arise from applying more restrictive thresholds is essential.
Collapse
Affiliation(s)
- Brenda Leeneman
- Department of Health Technology Assessment, Erasmus School of Health Policy and Management, Erasmus University Rotterdam, Burgemeester Oudlaan 50, 3062 PA Rotterdam, the Netherlands; Erasmus Centre for Health Economics Rotterdam, Erasmus University Rotterdam, Burgemeester Oudlaan 50, 3062 PA Rotterdam, the Netherlands.
| | - Nicolas S H Xander
- Department of Health Technology Assessment, Erasmus School of Health Policy and Management, Erasmus University Rotterdam, Burgemeester Oudlaan 50, 3062 PA Rotterdam, the Netherlands; Erasmus Centre for Health Economics Rotterdam, Erasmus University Rotterdam, Burgemeester Oudlaan 50, 3062 PA Rotterdam, the Netherlands
| | - W Edward Fiets
- Department of Medical Oncology, Medical Center Leeuwarden, Henri Dunantweg 2, 8934 AD Leeuwarden, the Netherlands
| | - Wouter K de Jong
- Department of Pulmonology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - Nathalie E M Uyl
- Department of Health Technology Assessment, Erasmus School of Health Policy and Management, Erasmus University Rotterdam, Burgemeester Oudlaan 50, 3062 PA Rotterdam, the Netherlands
| | - A N Machteld Wymenga
- Department of Medical Oncology, Medisch Spectrum Twente, Koningsplein 1, 7512 KZ Enschede the Netherlands
| | - An K L Reyners
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, the Netherlands
| | - Carin A Uyl-de Groot
- Department of Health Technology Assessment, Erasmus School of Health Policy and Management, Erasmus University Rotterdam, Burgemeester Oudlaan 50, 3062 PA Rotterdam, the Netherlands; Erasmus Centre for Health Economics Rotterdam, Erasmus University Rotterdam, Burgemeester Oudlaan 50, 3062 PA Rotterdam, the Netherlands
| |
Collapse
|
264
|
Wang K, Huang H, Zhan Q, Ding H, Li Y. Toll-like receptors in health and disease. MedComm (Beijing) 2024; 5:e549. [PMID: 38685971 PMCID: PMC11057423 DOI: 10.1002/mco2.549] [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: 07/31/2023] [Revised: 03/17/2024] [Accepted: 03/26/2024] [Indexed: 05/02/2024] Open
Abstract
Toll-like receptors (TLRs) are inflammatory triggers and belong to a family of pattern recognition receptors (PRRs) that are central to the regulation of host protective adaptive immune responses. Activation of TLRs in innate immune myeloid cells directs lymphocytes to produce the most appropriate effector responses to eliminate infection and maintain homeostasis of the body's internal environment. Inappropriate TLR stimulation can lead to the development of general autoimmune diseases as well as chronic and acute inflammation, and even cancer. Therefore, TLRs are expected to be targets for therapeutic treatment of inflammation-related diseases, autoimmune diseases, microbial infections, and human cancers. This review summarizes the recent discoveries in the molecular and structural biology of TLRs. The role of different TLR signaling pathways in inflammatory diseases, autoimmune diseases such as diabetes, cardiovascular diseases, respiratory diseases, digestive diseases, and even cancers (oral, gastric, breast, colorectal) is highlighted and summarizes new drugs and related clinical treatments in clinical trials, providing an overview of the potential and prospects of TLRs for the treatment of TLR-related diseases.
Collapse
Affiliation(s)
- Kunyu Wang
- Department of Head and Neck Oncology Surgery, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Hanyao Huang
- Department of Oral and Maxillofacial Surgery, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduSichuanChina
| | - Qi Zhan
- Department of Head and Neck Oncology Surgery, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Haoran Ding
- Department of Head and Neck Oncology Surgery, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Yi Li
- Department of Head and Neck Oncology Surgery, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| |
Collapse
|
265
|
Cook SL, Al Amin M, Bari S, Poonnen PJ, Khasraw M, Johnson MO. Immune Checkpoint Inhibitors in Geriatric Oncology. Curr Oncol Rep 2024; 26:562-572. [PMID: 38587598 DOI: 10.1007/s11912-024-01528-3] [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] [Accepted: 03/27/2024] [Indexed: 04/09/2024]
Abstract
PURPOSE OF REVIEW This manuscript will update prior reviews of immune checkpoint inhibitors (ICIs) in light of basic science, translational, and clinical discoveries in the field of cancer immunology and aging. RECENT FINDINGS ICIs have led to significant advancements in the treatment of cancer. Landmark trials of ICIs have cited the efficacy and toxicity experienced by older patients, but most trials are not specifically designed to address outcomes in older patients. Underlying mechanisms of aging, like cellular senescence, affect the immune system and may ultimately alter the host's response to ICIs. Validated tools are currently used to identify older adults who may be at greater risk of developing complications from their cancer treatment. We review changes in the aging immune system that may alter responses to ICIs, report outcomes and toxicities in older adults from recent ICI clinical trials, and discuss clinical tools specific to older patients with cancer.
Collapse
Affiliation(s)
- Sarah L Cook
- Department of Neurosurgery, Duke University School of Medicine, 047 Baker House, Trent Drive, Durham, NC, 27710, USA
| | - Md Al Amin
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
- Sher-E-Bangla Medical College, Barisal City, Bangladesh
| | - Shahla Bari
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Pradeep J Poonnen
- Department of Veterans Affairs, Durham VA Medical Center, Durham, NC, USA
- Department of Veterans Affairs, National TeleOncology Program, Durham, NC, USA
| | - Mustafa Khasraw
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA.
- Department of Neurosurgery, Duke University School of Medicine, 047 Baker House, Trent Drive, Durham, NC, 27710, USA.
| | - Margaret O Johnson
- Department of Neurosurgery, Duke University School of Medicine, 047 Baker House, Trent Drive, Durham, NC, 27710, USA
- Department of Veterans Affairs, National TeleOncology Program, Durham, NC, USA
| |
Collapse
|
266
|
Wu D, Yang L, Yan Y, Jiang Z, Liu Y, Dong P, Lv Y, Zhou S, Qiu Y, Yu X. Neoadjuvant immunotherapy improves outcomes for resectable gastroesophageal junction cancer: A systematic review and meta-analysis. Cancer Med 2024; 13:e7176. [PMID: 38716645 PMCID: PMC11077431 DOI: 10.1002/cam4.7176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 03/22/2024] [Accepted: 03/28/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND In recent years, neoadjuvant immunotherapy (NAIT) has developed rapidly in patients with gastroesophageal junction cancer (GEJC). The suggested neoadjuvant treatment regimens for patients with GEJC may vary in light of the efficacy and safety results. METHODS A search of the Cochrane Library, PubMed, Embase, and Web of Science was completed to locate studies examining the safety and effectiveness of NAIT for resectable GEJC. We analyzed the effect sizes (ES) and 95% confidence intervals (CI) in addition to subgroups and heterogeneity. Meta-analyses were performed using Stata BE17 software. RESULTS For these meta-analyses, 753 patients were chosen from 21 studies. The effectiveness of NAIT was assessed using the pathological complete response (pCR), major pathological response (MPR), and nodal downstage to ypN0 rate. The MPR, pCR, and nodal downstage to ypN0 rate values in NAIT were noticeably higher (MPR: ES = 0.45; 95% CI: 0.36-0.54; pCR: ES = 0.26; 95% CI: 0.21-0.32; nodal downstage to ypN0 rate: ES = 0.60; 95% CI: 0.48-0.72) than those of neoadjuvant chemotherapy (nCT) or neoadjuvant chemoradiotherapy (nCRT) (MPR < 30%; pCR: ES = 3%-17%; nodal downstage to ypN0 rate: ES = 21%-29%). Safety was assessed using the treatment-related adverse events (trAEs) incidence rate, surgical delay rate, surgical complications incidence rate, and surgical resection rate. In conclusion, the incidence of trAEs, incidence of surgical complications, and surgical delay rate had ES values of 0.66, 0.48, and 0.09, respectively. These rates were comparable to those from nCT or nCRT (95% CI: 0.60-0.70; 0.15-0.51; and 0, respectively). The reported resection rates of 85%-95% with nCT or nCRT were comparable to the mean surgical resection rate of 90%. CONCLUSION NAIT is an effective treatment for resectable GEJC; additionally, the level of NAIT toxicity is acceptable. The long-term effects of NAIT require further study.
Collapse
Affiliation(s)
- Danzhu Wu
- Clinical Medical College of Jining Medical UniversityJiningShandong ProvinceChina
| | - Liyuan Yang
- Shandong First Medical University and Shandong Academy of Medical SciencesJinanChina
| | - Yu Yan
- Shandong First Medical University and Shandong Academy of Medical SciencesJinanChina
| | - Zhengchen Jiang
- Department of General Surgery, Shandong Provincial Qianfoshan Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
| | - Yinglong Liu
- Shandong First Medical University and Shandong Academy of Medical SciencesJinanChina
| | - Peng Dong
- Department of OncologyThe Second Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, TaianJinanShandongChina
| | - Yajuan Lv
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer InstituteNational Key Laboratory of Advanced Drug Delivery and Release SystemsJinanShandongChina
| | - Siqin Zhou
- Medical CollegeWuhan University of Science and TechnologyWuhanChina
| | - Yiyang Qiu
- School of Nursing, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei ProvinceChina
| | - Xinshuang Yu
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer InstituteNational Key Laboratory of Advanced Drug Delivery and Release SystemsJinanShandongChina
| |
Collapse
|
267
|
de Haas AM, Stolk DA, Schetters STT, Goossens-Kruijssen L, Keuning E, Ambrosini M, Boon L, Kalay H, Storm G, van der Vliet HJ, de Gruijl TD, van Kooyk Y. Vaccination with DC-SIGN-Targeting αGC Liposomes Leads to Tumor Control, Irrespective of Suboptimally Activated T-Cells. Pharmaceutics 2024; 16:581. [PMID: 38794243 PMCID: PMC11124829 DOI: 10.3390/pharmaceutics16050581] [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: 03/19/2024] [Revised: 04/07/2024] [Accepted: 04/15/2024] [Indexed: 05/26/2024] Open
Abstract
Cancer vaccines have emerged as a potent strategy to improve cancer immunity, with or without the combination of checkpoint blockade. In our investigation, liposomal formulations containing synthetic long peptides and α-Galactosylceramide, along with a DC-SIGN-targeting ligand, Lewis Y (LeY), were studied for their anti-tumor potential. The formulated liposomes boosted with anti-CD40 adjuvant demonstrated robust invariant natural killer (iNKT), CD4+, and CD8+ T-cell activation in vivo. The incorporation of LeY facilitated the targeting of antigen-presenting cells expressing DC-SIGN in vitro and in vivo. Surprisingly, mice vaccinated with LeY-modified liposomes exhibited comparable tumor reduction and survival rates to those treated with untargeted counterparts despite a decrease in antigen-specific CD8+ T-cell responses. These results suggest that impaired induction of antigen-specific CD8+ T-cells via DC-SIGN targeting does not compromise anti-tumor potential, hinting at alternative immune activation routes beyond CD8+ T-cell activation.
Collapse
Affiliation(s)
- Aram M. de Haas
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Dorian A. Stolk
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Department of Medical Oncology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Sjoerd T. T. Schetters
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Laura Goossens-Kruijssen
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Eelco Keuning
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Martino Ambrosini
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- LIPOSOMA BV, Meerpaalweg 5, 1332 BB Almere, The Netherlands
| | | | - Hakan Kalay
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Gert Storm
- LIPOSOMA BV, Meerpaalweg 5, 1332 BB Almere, The Netherlands
- Department of Biomaterials Science and Technology, University of Twente, 7500 AE Enschede, The Netherlands
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Hans J. van der Vliet
- Department of Medical Oncology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- LAVA Therapeutics, 3584 CM Utrecht, The Netherlands
| | - Tanja D. de Gruijl
- Department of Medical Oncology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Yvette van Kooyk
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
268
|
Wu S, Dai X, Xia Y, Zhao Q, Zhao H, Shi Z, Yin X, Liu X, Zhang A, Yao Z, Zhang H, Li Q, Thorne RF, Zhang S, Sheng W, Hu W, Gu H. Targeting high circDNA2v levels in colorectal cancer induces cellular senescence and elicits an anti-tumor secretome. Cell Rep 2024; 43:114111. [PMID: 38615319 DOI: 10.1016/j.celrep.2024.114111] [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: 09/25/2023] [Revised: 02/03/2024] [Accepted: 03/28/2024] [Indexed: 04/16/2024] Open
Abstract
The efficacy of immunotherapy against colorectal cancer (CRC) is impaired by insufficient immune cell recruitment into the tumor microenvironment. Our study shows that targeting circDNA2v, a circular RNA commonly overexpressed in CRC, can be exploited to elicit cytotoxic T cell recruitment. circDNA2v functions through binding to IGF2BP3, preventing its ubiquitination, and prolonging the IGF2BP3 half-life, which in turn sustains mRNA levels of the protooncogene c-Myc. Targeting circDNA2v by gene silencing downregulates c-Myc to concordantly induce tumor cell senescence and the release of proinflammatory mediators. Production of CXCL10 and interleukin-9 by CRC cells is elicited through JAK-STAT1 signaling, in turn promoting the chemotactic and cytolytic activities of CD8+ T cells. Clinical evidence associates increased circDNA2v expression in CRC tissues with reductions in CD8+ T cell infiltration and worse outcomes. The regulatory relationship between circDNA2v, cellular senescence, and tumor-infiltrating lymphocytes thus provides a rational approach for improving immunotherapy in CRC.
Collapse
Affiliation(s)
- Shuang Wu
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Xiangyu Dai
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Yang Xia
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Qingsong Zhao
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Heng Zhao
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Zhimin Shi
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Xin Yin
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Xue Liu
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Aijie Zhang
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Zhihui Yao
- Translational Research Institute, People's Hospital of Zhengzhou University, Academy of Medical Science, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Tianjian Laboratory of Advanced Biomedical Sciences, State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou 450003, China
| | - Hao Zhang
- Department of Gastrointestinal Surgery, Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Qun Li
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Rick Francis Thorne
- Translational Research Institute, People's Hospital of Zhengzhou University, Academy of Medical Science, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Tianjian Laboratory of Advanced Biomedical Sciences, State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou 450003, China
| | - Shangxin Zhang
- Department of Gastrointestinal Surgery, Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Weiwei Sheng
- Department of Gastrointestinal Surgery, Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei 230022, China.
| | - Wanglai Hu
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China; Translational Research Institute, People's Hospital of Zhengzhou University, Academy of Medical Science, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Tianjian Laboratory of Advanced Biomedical Sciences, State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou 450003, China.
| | - Hao Gu
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
269
|
Fateeva A, Eddy K, Chen S. Current State of Melanoma Therapy and Next Steps: Battling Therapeutic Resistance. Cancers (Basel) 2024; 16:1571. [PMID: 38672652 PMCID: PMC11049326 DOI: 10.3390/cancers16081571] [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: 03/05/2024] [Revised: 04/11/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Melanoma is the most aggressive and deadly form of skin cancer due to its high propensity to metastasize to distant organs. Significant progress has been made in the last few decades in melanoma therapeutics, most notably in targeted therapy and immunotherapy. These approaches have greatly improved treatment response outcomes; however, they remain limited in their abilities to hinder disease progression due, in part, to the onset of acquired resistance. In parallel, intrinsic resistance to therapy remains an issue to be resolved. In this review, we summarize currently available therapeutic options for melanoma treatment and focus on possible mechanisms that drive therapeutic resistance. A better understanding of therapy resistance will provide improved rational strategies to overcome these obstacles.
Collapse
Affiliation(s)
- Anna Fateeva
- Susan Lehman Cullman Laboratory for Cancer Research, Rutgers University, Piscataway, NJ 08854, USA; (A.F.); (K.E.)
- Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, NJ 08854, USA
| | - Kevinn Eddy
- Susan Lehman Cullman Laboratory for Cancer Research, Rutgers University, Piscataway, NJ 08854, USA; (A.F.); (K.E.)
- Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, NJ 08854, USA
| | - Suzie Chen
- Susan Lehman Cullman Laboratory for Cancer Research, Rutgers University, Piscataway, NJ 08854, USA; (A.F.); (K.E.)
- Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, NJ 08854, USA
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA
- U.S. Department of Veterans Affairs, New Jersey Health Care System, East Orange, NJ 07018, USA
| |
Collapse
|
270
|
Crucitta S, Cucchiara F, Marconcini R, Bulleri A, Manacorda S, Capuano A, Cioni D, Nuzzo A, de Jonge E, Mathjissen RHJ, Neri E, van Schaik RHN, Fogli S, Danesi R, Del Re M. TGF-β mRNA levels in circulating extracellular vesicles are associated with response to anti-PD1 treatment in metastatic melanoma. Front Mol Biosci 2024; 11:1288677. [PMID: 38633217 PMCID: PMC11021649 DOI: 10.3389/fmolb.2024.1288677] [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: 09/04/2023] [Accepted: 02/27/2024] [Indexed: 04/19/2024] Open
Abstract
Introduction: Immune checkpoint inhibitors (ICIs) represent the standard therapy for metastatic melanoma. However, a few patients do not respond to ICIs and reliable predictive biomarkers are needed. Methods: This pilot study investigates the association between mRNA levels of programmed cell death-1 (PD-1) ligand 1 (PD-L1), interferon-gamma (IFN-γ), and transforming growth factor-β (TGF-β) in circulating extracellular vesicles (EVs) and survival in 30 patients with metastatic melanoma treated with first line anti-PD-1 antibodies. Blood samples were collected at baseline and RNA extracted from EVs; the RNA levels of PD-L1, IFN-γ, and TGF-β were analysed by digital droplet PCR (ddPCR). A biomarker-radiomic correlation analysis was performed in a subset of patients. Results: Patients with high TGF-β expression (cut-off fractional abundance [FA] >0.19) at baseline had longer median progression-free survival (8.4 vs. 1.8 months; p = 0.006) and overall survival (17.9 vs. 2.63 months; p = 0.0009). Moreover, radiomic analysis demonstrated that patients with high TGF-β expression at baseline had smaller lesions (2.41 ± 3.27 mL vs. 42.79 ± 101.08 mL, p < 0.001) and higher dissimilarity (12.01 ± 28.23 vs. 5.65 ± 8.4; p = 0.018). Discussion: These results provide evidence that high TGF-β expression in EVs is associated with a better response to immunotherapy. Further investigation on a larger patient population is needed to validate the predictive power of this potential biomarker of response to ICIs.
Collapse
Affiliation(s)
- Stefania Crucitta
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Federico Cucchiara
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Riccardo Marconcini
- Unit of Medical Oncology 2, Department of Medicine and Oncology, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Alessandra Bulleri
- Unit of Radiodiagnostics 1, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Simona Manacorda
- Unit of Medical Oncology 2, Department of Medicine and Oncology, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Annalisa Capuano
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Section of Pharmacology, Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Napoli, Italy
| | - Dania Cioni
- Unit of Radiodiagnostics 1, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Amedeo Nuzzo
- Unit of Medical Oncology 2, Department of Medicine and Oncology, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Evert de Jonge
- Department of Clinical Chemistry, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Ron H. J. Mathjissen
- Department of Medical Oncology, Erasmus University Medical Center Cancer Institute, Rotterdam, Netherlands
| | - Emanuele Neri
- Unit of Radiodiagnostics 1, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Ron H. N. van Schaik
- Department of Clinical Chemistry, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Stefano Fogli
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Romano Danesi
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
- Department of Oncology and Hemato-Oncology, University of Milano, Milano, Italy
| | - Marzia Del Re
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
271
|
El-Khoueiry AB, Trojan J, Meyer T, Yau T, Melero I, Kudo M, Hsu C, Kim TY, Choo SP, Kang YK, Yeo W, Chopra A, Soleymani S, Yao J, Neely J, Tschaika M, Welling TH, Sangro B. Nivolumab in sorafenib-naive and sorafenib-experienced patients with advanced hepatocellular carcinoma: 5-year follow-up from CheckMate 040. Ann Oncol 2024; 35:381-391. [PMID: 38151184 DOI: 10.1016/j.annonc.2023.12.008] [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: 05/30/2023] [Revised: 12/07/2023] [Accepted: 12/11/2023] [Indexed: 12/29/2023] Open
Abstract
BACKGROUND Patients with advanced hepatocellular carcinoma (aHCC) have a poor prognosis and high mortality. Nivolumab monotherapy demonstrated clinical benefit with an acceptable safety profile in patients with aHCC in the CheckMate 040 study. Five-year follow-up of the sorafenib-naive and sorafenib-experienced groups of CheckMate 040 is presented here. PATIENTS AND METHODS Patients received nivolumab monotherapy at dose levels of 0.1-10.0 mg/kg (dose-escalation phase) or 3 mg/kg (dose-expansion phase) every 2 weeks until disease progression or unacceptable toxicity. Primary endpoints were safety and tolerability (dose escalation), and objective response rate (ORR) by blinded independent central review (BICR) and by investigator as per RECIST version 1.1 (dose expansion). RESULTS Eighty sorafenib-naive and 154 sorafenib-experienced patients were treated. Minimum follow-up in both groups was 60 months. ORR as per BICR was 20% [95% confidence interval (CI) 12% to 30%] and 14% (95% CI 9% to 21%) in the sorafenib-naive and sorafenib-experienced groups, respectively. Responses occurred regardless of HCC etiology or baseline tumor cell programmed death-ligand 1 (PD-L1) expression levels. Median overall survival (OS) was 26.6 months (95% CI 16.6-30.6 months) and 15.1 months (95% CI 13.0-18.2 months) in sorafenib-naive and sorafenib-experienced patients, respectively. The 3-year OS rates were 28% in the sorafenib-naive and 20% in the sorafenib-experienced groups; 5-year OS rates were 14% and 12%, respectively. No new safety signals were identified; grade 3/4 treatment-related adverse events were observed in 33% and 21% of patients in the sorafenib-naive and sorafenib-experienced groups, respectively. Biomarker analyses showed that baseline PD-L1 expression ≥1% was associated with higher ORR and longer OS compared with PD-L1 <1%. In the sorafenib-naive group, patients with OS ≥3 years exhibited higher baseline CD8 T-cell density compared with those with OS <1 year. CONCLUSION With 5 years of follow-up, nivolumab monotherapy continued to provide durable clinical benefit with manageable safety in sorafenib-naive and sorafenib-experienced patients with aHCC.
Collapse
Affiliation(s)
- A B El-Khoueiry
- Division of Medical Oncology, USC Norris Comprehensive Cancer Center, Los Angeles, USA.
| | - J Trojan
- Department of Medicine, Goethe University Hospital and Cancer Center, Frankfurt, Germany
| | - T Meyer
- Department of Oncology, Royal Free Hospital, London, UK
| | - T Yau
- Department of Medicine, University of Hong Kong, Hong Kong, China
| | - I Melero
- Department of Immunology, Clinica Universidad de Navarra and CIBERONC, Pamplona, Spain
| | - M Kudo
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka, Japan
| | - C Hsu
- Department of Medical Oncology, National Taiwan University Cancer Center, Taipei, Taiwan; Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - T-Y Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - S-P Choo
- Division of Medical Oncology, National Cancer Center and Curie Oncology, Singapore, Republic of Singapore
| | - Y-K Kang
- Department of Oncology, Asan Medical Center, University of Ulsan, Seoul, Korea
| | - W Yeo
- Department of Clinical Oncology, Chinese University of Hong Kong, Hong Kong, China
| | - A Chopra
- Department of Medical Oncology, Johns Hopkins Singapore International Medical Centre, Singapore, Republic of Singapore
| | - S Soleymani
- Global Biometrics & Data Sciences, Bristol Myers Squibb, Princeton, USA
| | - J Yao
- Informatics and Predictive Sciences, Bristol Myers Squibb, Princeton, USA
| | - J Neely
- Translational Medicine, Bristol Myers Squibb, Princeton, USA
| | - M Tschaika
- Oncology Clinical Development, Bristol Myers Squibb, Princeton, USA
| | - T H Welling
- Perlmutter Cancer Center and Department of Surgery, NYU Langone Health, New York, USA
| | - B Sangro
- Liver Unit and HPB Oncology Area, Clinica Universidad de Navarra and CIBEREHD, Pamplona, Spain
| |
Collapse
|
272
|
Debono J, Balzan D, Borg JJ, Falzon S, al-Haddad D, Micallef B, Sultana J. Nivolumab Safety in Renal Cell Carcinoma: A Case Report. J Pharm Technol 2024; 40:112-117. [PMID: 38525093 PMCID: PMC10959086 DOI: 10.1177/87551225231218164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2024] Open
Abstract
Nivolumab is used to treat several different types of cancers. Although it is generally considered to be effective and well-tolerated, it has been associated with adverse effects requiring discontinuation of treatment, like many other drugs used for cancer. A 70-year-old male was switched from sunitinib to nivolumab for renal cell carcinoma. The patient developed persistent hypothyroidism, onycholysis, and pneumonitis at nivolumab cycle 6, 10, and 11, respectively. Using the Naranjo causality method, the likelihood of causality was deemed "probable" for pneumonitis and hypothyroidism and "possible" for onycholysis. Nivolumab was eventually discontinued due to disease progression, rather than safety concerns. Eudravigilance, the European pharmacovigilance database, was searched for all nivolumab-related individual case safety reports from Malta, up to September 4, 2023. Six reports were identified in Malta, although the 3 events identified in this case report were not reported, suggesting under-reporting in Malta. This case report identified an uncommon nivolumab adverse drug reaction (ADR), onycholysis and showed how, despite the occurrence of 3 ADRs, it was its lack of efficacy rather than its safety which led to its discontinuation in this particular patient.
Collapse
Affiliation(s)
- James Debono
- Oncology Department, Sir Anthony Mamo Oncology Centre, Mater Dei Hospital, Msida, Malta
| | - Dustin Balzan
- Pharmacy Directorate, Mater Dei Hospital, Msida, Malta
| | | | | | | | | | - Janet Sultana
- Pharmacy Directorate, Mater Dei Hospital, Msida, Malta
- Exeter College of Medicine and Health, University of Exeter, Exeter, UK
| |
Collapse
|
273
|
Hayashi H, Chamoto K, Hatae R, Kurosaki T, Togashi Y, Fukuoka K, Goto M, Chiba Y, Tomida S, Ota T, Haratani K, Takahama T, Tanizaki J, Yoshida T, Iwasa T, Tanaka K, Takeda M, Hirano T, Yoshida H, Ozasa H, Sakamori Y, Sakai K, Higuchi K, Uga H, Suminaka C, Hirai T, Nishio K, Nakagawa K, Honjo T. Soluble immune checkpoint factors reflect exhaustion of antitumor immunity and response to PD-1 blockade. J Clin Invest 2024; 134:e168318. [PMID: 38557498 PMCID: PMC10977985 DOI: 10.1172/jci168318] [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: 12/22/2022] [Accepted: 01/30/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUNDPrecise stratification of patients with non-small cell lung cancer (NSCLC) is needed for appropriate application of PD-1/PD-L1 blockade therapy.METHODSWe measured soluble forms of the immune-checkpoint molecules PD-L1, PD-1, and CTLA-4 in plasma of patients with advanced NSCLC before PD-1/PD-L1 blockade. A prospective biomarker-finding trial (cohort A) included 50 previously treated patients who received nivolumab. A retrospective observational study was performed for patients treated with any PD-1/PD-L1 blockade therapy (cohorts B and C), cytotoxic chemotherapy (cohort D), or targeted therapy (cohort E). Plasma samples from all patients were assayed for soluble immune-checkpoint molecules with a highly sensitive chemiluminescence-based assay.RESULTSNonresponsiveness to PD-1/PD-L1 blockade therapy was associated with higher concentrations of these soluble immune factors among patients with immune-reactive (hot) tumors. Such an association was not apparent for patients treated with cytotoxic chemotherapy or targeted therapy. Integrative analysis of tumor size, PD-L1 expression in tumor tissue (tPD-L1), and gene expression in tumor tissue and peripheral CD8+ T cells revealed that high concentrations of the 3 soluble immune factors were associated with hyper or terminal exhaustion of antitumor immunity. The combination of soluble PD-L1 (sPD-L1) and sCTLA-4 efficiently discriminated responsiveness to PD-1/PD-L1 blockade among patients with immune-reactive tumors.CONCLUSIONCombinations of soluble immune factors might be able to identify patients unlikely to respond to PD-1/PD-L1 blockade as a result of terminal exhaustion of antitumor immunity. Our data suggest that such a combination better predicts, along with tPD-L1, for the response of patients with NSCLC.TRIAL REGISTRATIONUMIN000019674.FUNDINGThis study was funded by Ono Pharmaceutical Co. Ltd. and Sysmex Corporation.
Collapse
Affiliation(s)
- Hidetoshi Hayashi
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Kenji Chamoto
- Department of Immunology and Genomic Medicine, Center for Cancer Immunotherapy and Immunobiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department of Immuno-Oncology PDT, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ryusuke Hatae
- Department of Immunology and Genomic Medicine, Center for Cancer Immunotherapy and Immunobiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takashi Kurosaki
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Yosuke Togashi
- Department of Genome Biology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
- Department of Tumor Microenvironment, Faculty of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Kazuya Fukuoka
- Clinical Research Center, Kindai University Hospital, Osaka-Sayama, Japan
| | | | - Yasutaka Chiba
- Clinical Research Center, Kindai University Hospital, Osaka-Sayama, Japan
| | - Shuta Tomida
- Department of Center for Comprehensive Genomic Medicine, Okayama University Hospital, Okayama, Japan
| | - Takayo Ota
- Department of Medical Oncology, Izumi City General Hospital, Izumi, Japan
| | - Koji Haratani
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Takayuki Takahama
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Junko Tanizaki
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Takeshi Yoshida
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Tsutomu Iwasa
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Kaoru Tanaka
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Masayuki Takeda
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
- Department of Cancer Genomics and Medical Oncology, Nara Medical University School of Medicine, Nara, Japan
| | - Tomoko Hirano
- Department of Immunology and Genomic Medicine, Center for Cancer Immunotherapy and Immunobiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hironori Yoshida
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroaki Ozasa
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yuichi Sakamori
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Kazuko Sakai
- Department of Genome Biology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | | | | | | | - Toyohiro Hirai
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kazuto Nishio
- Department of Genome Biology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Kazuhiko Nakagawa
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Tasuku Honjo
- Department of Immunology and Genomic Medicine, Center for Cancer Immunotherapy and Immunobiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
274
|
Boutros A, Croce E, Ferrari M, Gili R, Massaro G, Marconcini R, Arecco L, Tanda ET, Spagnolo F. The treatment of advanced melanoma: Current approaches and new challenges. Crit Rev Oncol Hematol 2024; 196:104276. [PMID: 38295889 DOI: 10.1016/j.critrevonc.2024.104276] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 01/15/2024] [Accepted: 01/26/2024] [Indexed: 02/17/2024] Open
Abstract
In recent years, advances in melanoma treatment have renewed patient hope. This comprehensive review emphasizes the evolving treatment landscape, particularly highlighting first-line strategies and the interplay between immune-checkpoint inhibitors (ICIs) and targeted therapies. Ipilimumab plus nivolumab has achieved the best median overall survival, exceeding 70 months. However, the introduction of new ICIs, like relatlimab, has added complexity to first-line therapy decisions. Our aim is to guide clinicians in making personalized treatment decisions. Various features, including brain metastases, PD-L1 expression, BRAF mutation, performance status, and prior adjuvant therapy, significantly impact the direction of advanced melanoma treatment. We also provide the latest insights into the treatment of rare melanoma subtypes, such as uveal melanoma, where tebentafusp has shown promising improvements in overall survival for metastatic uveal melanoma patients. This review provides invaluable insights for clinicians, enabling informed treatment choices and deepening our understanding of the multifaceted challenges associated with advanced melanoma management.
Collapse
Affiliation(s)
- Andrea Boutros
- Skin Cancer Unit, U.O. Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, Genova, Italy; Department of Internal Medicine and Medical Sciences (DiMI), School of Medicine, University of Genova, Genova, Italy.
| | - Elena Croce
- Skin Cancer Unit, U.O. Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Marco Ferrari
- Azienda Ospedaliero Universitaria Pisana, Medical Oncology Unit, Pisa, Italy
| | - Riccardo Gili
- Skin Cancer Unit, U.O. Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, Genova, Italy; Department of Internal Medicine and Medical Sciences (DiMI), School of Medicine, University of Genova, Genova, Italy
| | - Giulia Massaro
- Unit of Medical Oncology, Careggi University-Hospital, 50134 Florence, Italy
| | - Riccardo Marconcini
- Azienda Ospedaliero Universitaria Pisana, Medical Oncology Unit, Pisa, Italy
| | - Luca Arecco
- Department of Internal Medicine and Medical Sciences (DiMI), School of Medicine, University of Genova, Genova, Italy; Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Enrica Teresa Tanda
- Skin Cancer Unit, U.O. Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Francesco Spagnolo
- Skin Cancer Unit, U.O. Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, Genova, Italy; Department of Surgical Sciences and Integrated Diagnostics (DISC), Plastic Surgery Division, University of Genova, Genova, Italy
| |
Collapse
|
275
|
Levi S, Bank H, Mullinax J, Boland G. Precision Oncology in Melanoma and Skin Cancer Surgery. Surg Oncol Clin N Am 2024; 33:369-385. [PMID: 38401915 DOI: 10.1016/j.soc.2023.12.017] [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] [Indexed: 02/26/2024]
Abstract
There has been perhaps no greater advance in the prognosis of solid tumors in the last decade than for patients with metastatic melanoma. This is due to significant improvements in treatment based on two key components of melanoma tumor biology (1) the identification of driver mutations with therapeutic potential and (2) the mechanistic understanding of a tumor-specific immune response. With breakthrough findings in such a relatively short period of time, the treatment of patients with metastatic melanoma has become intensely personalized.
Collapse
Affiliation(s)
| | | | - John Mullinax
- Sarcoma Department, Moffitt Cancer Center, Tampa, FL, USA
| | - Genevieve Boland
- Department of Surgery, MGH, Boston, MA, USA; Department of Surgery, Massachusetts General Hospital (MGH) Cancer Center, Harvard Medical School (HMS), Boston, MA, USA.
| |
Collapse
|
276
|
Stassen RC, Maas CCHM, van der Veldt AAM, Lo SN, Saw RPM, Varey AHR, Scolyer RA, Long GV, Thompson JF, Rutkowski P, Keilholz U, van Akkooi ACJ, Verhoef C, van Klaveren D, Grünhagen DJ. Development and validation of a novel model to predict recurrence-free survival and melanoma-specific survival after sentinel lymph node biopsy in patients with melanoma: an international, retrospective, multicentre analysis. Lancet Oncol 2024; 25:509-517. [PMID: 38547894 DOI: 10.1016/s1470-2045(24)00076-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 01/19/2024] [Accepted: 01/30/2024] [Indexed: 04/02/2024]
Abstract
BACKGROUND The introduction of adjuvant systemic treatment for patients with high-risk melanomas necessitates accurate staging of disease. However, inconsistencies in outcomes exist between disease stages as defined by the American Joint Committee on Cancer (8th edition). We aimed to develop a tool to predict patient-specific outcomes in people with melanoma rather than grouping patients according to disease stage. METHODS Patients older than 13 years with confirmed primary melanoma who underwent sentinel lymph node biopsy (SLNB) between Oct 29, 1997, and Nov 11, 2013, at four European melanoma centres (based in Berlin, Germany; Amsterdam and Rotterdam, the Netherlands; and Warsaw, Poland) were included in the development cohort. Potential predictors of recurrence-free and melanoma-specific survival assessed were sex, age, presence of ulceration, primary tumour location, histological subtype, Breslow thickness, sentinel node status, number of sentinel nodes removed, maximum diameter of the largest sentinel node metastasis, and Dewar classification. A prognostic model and nomogram were developed to predict 5-year recurrence-free survival on a continuous scale in patients with stage pT1b or higher melanomas. This model was also calibrated to predict melanoma-specific survival. Model performance was assessed by discrimination (area under the time-dependent receiver operating characteristics curve [AUC]) and calibration. External validation was done in a cohort of patients with primary melanomas who underwent SLNB between Jan 30, 1997, and Dec 12, 2013, at the Melanoma Institute Australia (Sydney, NSW, Australia). FINDINGS The development cohort consisted of 4071 patients, of whom 2075 (51%) were female and 1996 (49%) were male. 889 (22%) had sentinel node-positive disease and 3182 (78%) had sentinel node-negative disease. The validation cohort comprised 4822 patients, of whom 1965 (41%) were female and 2857 (59%) were male. 891 (18%) had sentinel node-positive disease and 3931 (82%) had sentinel node-negative disease. Median follow-up was 4·8 years (IQR 2·3-7·8) in the development cohort and 5·0 years (2·2-8·9) in the validation cohort. In the development cohort, 5-year recurrence-free survival was 73·5% (95% CI 72·0-75·1) and 5-year melanoma-specific survival was 86·5% (85·3-87·8). In the validation cohort, the corresponding estimates were 66·1% (64·6-67·7) and 83·3% (82·0-84·6), respectively. The final model contained six prognostic factors: sentinel node status, Breslow thickness, presence of ulceration, age at SLNB, primary tumour location, and maximum diameter of the largest sentinel node metastasis. In the development cohort, for the model's prediction of recurrence-free survival, the AUC was 0·80 (95% CI 0·78-0·81); for prediction of melanoma-specific survival, the AUC was 0·81 (0·79-0·84). External validation showed good calibration for both outcomes, with AUCs of 0·73 (0·71-0·75) and 0·76 (0·74-0·78), respectively. INTERPRETATION Our prediction model and nomogram accurately predicted patient-specific risk probabilities for 5-year recurrence-free and melanoma-specific survival. These tools could have important implications for clinical decision making when considering adjuvant treatments in patients with high-risk melanomas. FUNDING Erasmus Medical Centre Cancer Institute.
Collapse
Affiliation(s)
- Robert C Stassen
- Department of Surgical Oncology, Erasmus Medical Centre Cancer Institute, Rotterdam, Netherlands
| | - Carolien C H M Maas
- Department of Public Health, Erasmus University Medical Centre, Rotterdam, Netherlands
| | - Astrid A M van der Veldt
- Department of Medical Oncology, Erasmus Medical Centre Cancer Institute, Rotterdam, Netherlands; Department of Radiology and Nuclear Medicine, Erasmus Medical Centre Cancer Institute, Rotterdam, Netherlands
| | - Serigne N Lo
- Melanoma Institute Australia, University of Sydney, Sydney, NSW, Australia; Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Robyn P M Saw
- Melanoma Institute Australia, University of Sydney, Sydney, NSW, Australia; Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Alexander H R Varey
- Melanoma Institute Australia, University of Sydney, Sydney, NSW, Australia; Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Department of Plastic Surgery, Westmead Hospital, Sydney, NSW, Australia
| | - Richard A Scolyer
- Melanoma Institute Australia, University of Sydney, Sydney, NSW, Australia; Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia; Department of Tissue Oncology and Diagnostic Pathology, Royal Prince Alfred Hospital, Sydney, NSW, Australia; Department of Tissue Oncology and Diagnostic Pathology, NSW Health Pathology, Sydney, NSW, Australia
| | - Georgina V Long
- Melanoma Institute Australia, University of Sydney, Sydney, NSW, Australia; Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia; Department of Medical Oncology, Royal North Shore Hospital and Mater Hospital, Sydney, NSW, Australia
| | - John F Thompson
- Melanoma Institute Australia, University of Sydney, Sydney, NSW, Australia; Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Skłodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Ulrich Keilholz
- Department of Haemato-oncology, Charité Universitätsmedizin, Berlin, Germany
| | - Alexander C J van Akkooi
- Melanoma Institute Australia, University of Sydney, Sydney, NSW, Australia; Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Cornelis Verhoef
- Department of Surgical Oncology, Erasmus Medical Centre Cancer Institute, Rotterdam, Netherlands
| | - David van Klaveren
- Department of Public Health, Erasmus University Medical Centre, Rotterdam, Netherlands
| | - Dirk J Grünhagen
- Department of Surgical Oncology, Erasmus Medical Centre Cancer Institute, Rotterdam, Netherlands.
| |
Collapse
|
277
|
Hori Y, Naitoh I, Naiki-Ito A, Kawai T, Yoshida M, Kato A, Kachi K, Sahashi H, Adachi A, Toyohara T, Kito Y, Yamamoto T, Takahashi S, Kataoka H. Incidence of Pancreatic Injury and Pancreatitis in Patients Treated With Immune Checkpoint Inhibitors. Clin Transl Gastroenterol 2024; 15:e00667. [PMID: 38088372 PMCID: PMC11042772 DOI: 10.14309/ctg.0000000000000667] [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: 10/12/2023] [Accepted: 12/07/2023] [Indexed: 04/26/2024] Open
Abstract
INTRODUCTION Immune checkpoint inhibitors (ICIs) are being increasingly used to treat advanced malignancies. ICI-induced pancreatic injury (ICI-PI), which is an immune-related adverse event that may be a risk factor of ICI-associated pancreatitis, is not well documented in the literature. METHODS Consecutive patients who received ICIs for advanced malignancies from August 2015 through October 2022 were analyzed for the incidence of ICI-PI based on the Common Terminology Criteria for Adverse Events and ICI-associated pancreatitis. The imaging, clinical, and pathological findings of ICI-associated pancreatitis were also assessed. RESULTS This study enrolled 843 patients. In multivariable analyses, dual or simultaneous immunotherapy and ≥10 cycles of ICI administration were significant predictive factors for all grades of pancreatic injury, including grade ≥3. Notably, patients who received simultaneous immunotherapy exhibited a higher incidence of grade ≥3 pancreatic injuries compared with those receiving asynchronous immunotherapy in univariable analysis ( P = 0.032). One-fifth of the patients (16/70) with grade ≥3 pancreatic injuries had imaging evidence of pancreatitis similar to mild acute pancreatitis. ICI-associated pancreatitis was observed in 5.7% (48/843) of patients, including 1.8% (15/843) with moderate-to-severe pancreatitis (grade ≥2). Symptomatic cases (0.36%, 3/843) were treated with steroids with favorable outcomes. Immunohistochemistry for CD4 and CD8 revealed greater infiltration of CD8 + than CD4 + lymphocytes. DISCUSSION Simultaneous immunotherapy and dual immunotherapy are risk factors of ICI-PI. Although most patients diagnosed with ICI-PI and ICI-associated pancreatitis were asymptomatic and had a low mortality likelihood, long-term outcomes, including endocrine and exocrine function, should be carefully monitored.
Collapse
Affiliation(s)
- Yasuki Hori
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Itaru Naitoh
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Aya Naiki-Ito
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Tatsuya Kawai
- Department of Radiology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
- Department of Radiology, Nagoya City University Midori Municipal Hospital, Nagoya, Japan
| | - Michihiro Yoshida
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Akihisa Kato
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Kenta Kachi
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Hidenori Sahashi
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Akihisa Adachi
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Tadashi Toyohara
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yusuke Kito
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Tatsuhito Yamamoto
- Department of Radiology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Satoru Takahashi
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Hiromi Kataoka
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| |
Collapse
|
278
|
Lee ST, Kovaleva N, Senko C, Kee D, Scott AM. Positron Emission Tomography/Computed Tomography Transformation of Oncology: Melanoma and Skin Malignancies. PET Clin 2024; 19:231-248. [PMID: 38233284 DOI: 10.1016/j.cpet.2023.12.009] [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] [Indexed: 01/19/2024]
Abstract
Skin cancers are the most common cancers, with melanoma resulting in the highest cause of death in this category. Accurate clinical, histologic, and imaging staging with fludeoxyglucose positron emission tomography (FDG PET) is most important to guide patient management. Whilst surgical excision with clear margins is the gold-standard treatment for primary cutaneous melanoma, targeted therapies have generated remarkable and rapid clinical responses in melanoma, for which FDG PET also plays an important role in assessment of treatment response and post-therapy surveillance. Non-FDG PET tracers, advanced PET technology, and PET radiomics may potentially change the landscape of the utilization of PET in the imaging of patients with cutaneous malignancies.
Collapse
Affiliation(s)
- Sze-Ting Lee
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, Australia; Department of Medicine, University of Melbourne, Melbourne, Australia; Olivia Newton-John Cancer Research Institute, and La Trobe University, Heidelberg, Australia; Department of Surgery, University of Melbourne, Melbourne, Australia; School of Health and Biomedical Sciences, RMIT University, Melbourne, Australia
| | - Natalia Kovaleva
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, Australia
| | - Clare Senko
- Olivia Newton-John Cancer Research Institute, and La Trobe University, Heidelberg, Australia; Department of Medical Oncology, Olivia Newton-John Cancer and Wellness Centre, Austin Health, Heidelberg, Australia
| | - Damien Kee
- Olivia Newton-John Cancer Research Institute, and La Trobe University, Heidelberg, Australia; Department of Medical Oncology, Olivia Newton-John Cancer and Wellness Centre, Austin Health, Heidelberg, Australia; Department of Medical Oncology, Peter MacCallum Cancer Center, Melbourne, Australia
| | - Andrew M Scott
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, Australia; Department of Medicine, University of Melbourne, Melbourne, Australia; Olivia Newton-John Cancer Research Institute, and La Trobe University, Heidelberg, Australia.
| |
Collapse
|
279
|
Baandrup L, Sand FL, Aalborg GL, Nøttrup TJ, Fiehn AMK, Kjaer SK. PD-L1 expression in vulvar cancer: a systematic review and meta-analysis. Histopathology 2024; 84:742-752. [PMID: 38084642 DOI: 10.1111/his.15112] [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: 10/11/2023] [Revised: 11/11/2023] [Accepted: 11/18/2023] [Indexed: 03/06/2024]
Abstract
Programmed cell death ligand-1 (PD-L1) expression in cancer may predict clinical response to immunotherapeutic treatment with PD-1/PD-L1 inhibitors. Within the vulvar cancer field, PD-L1 expression has only been assessed by a few studies. We conducted a meta-analysis to examine the prevalence of PD-L1 positivity in vulvar cancer. PubMed, Embase, and Cochrane were searched for articles reporting on PD-L1 expression in vulvar cancer. Study selection and data extraction were performed independently by two authors. We extracted data on PD-L1 prevalence in vulvar cancer according to combined positive score (CPS) and tumour proportion score (TPS). Cutoff values for positivity were ≥1 or ≥10 for CPS and ≥1% and ≥5% for TPS. Random-effects models were used to estimate pooled PD-L1 prevalence, with 95% confidence intervals (CIs). Tests of between-study heterogeneity were evaluated by the I2 statistics. Sources of heterogeneity were explored by subgroup analyses and meta-regression. In total, 19 studies were included. Pooled PD-L1 prevalence in vulvar cancer was 83.4% (95% CI: 70.8-91.3; I2 = 80.0) and 53.9% (95% CI: 37.4-69.6; I2 = 93.0) according to CPS and TPS, respectively. Based on TPS, human papillomavirus (HPV)-associated vulvar squamous cell carcinomas (SCC) showed a lower PD-L1 prevalence (39.9%; 95% CI: 13.3-74.2) compared with HPV-independent SCC (62.6%; 95% CI: 33.7-84.6), but meta-regression showed no significant variation in PD-L1 prevalence by HPV status. PD-L1 prevalence was similar in advanced (44.9%; 95% CI: 29.8-61.1) and localized vulvar cancer (56.7%; 95% CI: 18.9-76.7). In conclusion, PD-L1 expression in vulvar cancer is frequent but between-study heterogeneity was high. Based on a subgroup of heterogenous studies, we found no strong variation in PD-L1 prevalence according to HPV status and stage.
Collapse
Affiliation(s)
- Louise Baandrup
- Unit of Virus, Lifestyle and Genes, Danish Cancer Institute, Copenhagen, Denmark
- Department of Pathology, Zealand University Hospital, Roskilde, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Freja Laerke Sand
- Unit of Virus, Lifestyle and Genes, Danish Cancer Institute, Copenhagen, Denmark
| | | | - Trine J Nøttrup
- Department of Oncology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Anne-Marie K Fiehn
- Department of Pathology, Zealand University Hospital, Roskilde, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Susanne K Kjaer
- Unit of Virus, Lifestyle and Genes, Danish Cancer Institute, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Juliane Marie Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
280
|
Bafaloukos D, Kouzis P, Gouveris P, Boukovinas I, Kalbakis K, Baka S, Kyriakakis G, Moschou D, Molfeta A, Demiri S, Mavroudis D, Spanoudi F, Dimitriadis I, Gogas H. Real-world management practices and characteristics of patients with advanced melanoma initiated on immuno-oncology or targeted therapy in the first-line setting during the period 2015-2018 in Greece. The 'SUMMER' study: a retrospective multicenter chart review project. Melanoma Res 2024; 34:152-165. [PMID: 38092014 PMCID: PMC10906211 DOI: 10.1097/cmr.0000000000000949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 11/03/2023] [Indexed: 02/02/2024]
Abstract
This study primarily aimed to generate real-world evidence (RWE) on the profile and first-line treatment (1LT) patterns of patients with advanced (unresectable Stage III/metastatic) cutaneous melanoma initiated on immuno-oncology (IO)- or targeted therapy (TT)-based 1LT between 1 January 2015 and 1 January 2018 (index period), in routine settings of Greece. This was a multicenter, retrospective chart review study. Eligible consented (unless deceased, for whom consent was waived by the hospital) patients were consecutively included by six oncology clinics. The look-back period extended from informed consent or death to initial melanoma diagnosis. Between 9 Junuary 2021 and 9 February 2022, 225 eligible patients (all Caucasians; 60.4% male; 35.6% diagnosed with de novo advanced melanoma) were included. At 1LT initiation, median age was 62.6 years; 2.7/6.7/90.7% of the patients had Stage IIIB/IIIC/IV disease and 9.3% were unresected. Most frequent metastatic sites were the lung (46.7%), non-regional nodes (33.8%), and liver (20.9%). Among patients, 98.2% had single primary melanoma, 45.6% had disease localized on the trunk, and 63.6% were BRAF-mutant. Of the patients, 45.3% initiated 1LT with an IO-based, 53.3% with a TT-based regimen, and three patients (1.3%) received TT-based followed by IO-based or vice versa. Most common 1LT patterns (frequency ≥10%) were BRAFi/MEKi combination (31.6%), anti-PD-1 monotherapy (25.3%), BRAFi monotherapy (21.8%), and anti-CTLA-4 monotherapy (17.8%). Most frequent regimens were Dabrafenib+Trametinib in 25.3%, and monotherapies with Pembrolizumab/Ipilimumab/Vemurafenib/Dabrafenib in 23.6/17.8/11.1/10.7% of patients, respectively. SUMMER provides RWE on 1LT strategies and profile of patients initiated 1L IO- or TT-based therapy in Greece during the 3-year index period.
Collapse
Affiliation(s)
| | - Panagiotis Kouzis
- First Department of Medicine, Laiko General Hospital, National and Kapodistrian University of Athens School of Medicine
| | | | | | | | - Sofia Baka
- Oncology Department, Interbalkan European Medical Center, Thessaloniki
| | - Georgios Kyriakakis
- First Department of Medicine, Laiko General Hospital, National and Kapodistrian University of Athens School of Medicine
| | - Despoina Moschou
- First Department of Medicine, Laiko General Hospital, National and Kapodistrian University of Athens School of Medicine
| | | | - Stamatia Demiri
- Second Department of Medical Oncology, Agios Savvas Hospital, Athens
| | - Dimitrios Mavroudis
- Department of Medical Oncology, University General Hospital of Heraklion, Heraklion
| | - Filio Spanoudi
- MSD Pharmaceutical, Industrial and Commercial S.A., Medical Affairs, Athens, Greece
| | - Ioannis Dimitriadis
- MSD Pharmaceutical, Industrial and Commercial S.A., Medical Affairs, Athens, Greece
| | - Helen Gogas
- First Department of Medicine, Laiko General Hospital, National and Kapodistrian University of Athens School of Medicine
| |
Collapse
|
281
|
Sobczuk P, Cholewiński M, Rutkowski P. Recent advances in tyrosine kinase inhibitors VEGFR 1-3 for the treatment of advanced metastatic melanoma. Expert Opin Pharmacother 2024; 25:501-510. [PMID: 38607407 DOI: 10.1080/14656566.2024.2342403] [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: 02/04/2024] [Accepted: 04/09/2024] [Indexed: 04/13/2024]
Abstract
INTRODUCTION Increasing evidence from preclinical and clinical studies suggests the role of vascular endothelial growth factor (VEGF) signaling in melanoma progression, response to therapy, and overall survival. Moreover, the discovery of the potential involvement of the VEGF pathway in resistance to immunotherapy has led to new clinical trials with VEGFR inhibitors. AREAS COVERED We have reviewed recent literature, mainly published within the last 5 years, on VEGFR-targeted treatments for advanced melanoma, including mucosal, acral, and uveal melanoma. The VEGFR inhibitors were used as a single therapy or combined with either immunotherapy or chemotherapy, and they were employed in treatment for KIT-mutated cutaneous melanoma and for patients with brain metastases. EXPERT OPINION Trials involving monotherapy have been unsuccessful in demonstrating meaningful efficacy. Despite some activity, the combination of VEGFR-targeting tyrosine kinase inhibitors (TKIs) with immune checkpoint inhibitors (ICI) in patients with ICI-resistant melanoma, the combination did not significantly improve outcomes compared to anti-PD-1 monotherapy in the first-line settings. On the contrary, some patients with mucosal, acral or KIT-mutant melanoma may benefit from TKI-based therapies. Further studies focused on biomarker discovery and randomized trials are necessary to better understand the role of VEGFR1-3 as a therapeutic target in melanoma.
Collapse
Affiliation(s)
- Paweł Sobczuk
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Skłodowska-Curie National Research institute of Oncology in Warsaw, Warsaw, Poland
| | - Michał Cholewiński
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Skłodowska-Curie National Research institute of Oncology in Warsaw, Warsaw, Poland
- Faculty of Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Skłodowska-Curie National Research institute of Oncology in Warsaw, Warsaw, Poland
| |
Collapse
|
282
|
Ruggiu M, Guérin MV, Corre B, Bardou M, Alonso R, Russo E, Garcia Z, Feldmann L, Lemaître F, Dusseaux M, Grandjean CL, Bousso P. Anti-PD-1 therapy triggers Tfh cell-dependent IL-4 release to boost CD8 T cell responses in tumor-draining lymph nodes. J Exp Med 2024; 221:e20232104. [PMID: 38417020 PMCID: PMC10901238 DOI: 10.1084/jem.20232104] [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: 11/15/2023] [Revised: 12/22/2023] [Accepted: 02/02/2024] [Indexed: 03/01/2024] Open
Abstract
Anti-PD-1 therapy targets intratumoral CD8+ T cells to promote clinical responses in cancer patients. Recent evidence suggests an additional activity in the periphery, but the underlying mechanism is unclear. Here, we show that anti-PD-1 mAb enhances CD8+ T cell responses in tumor-draining lymph nodes by stimulating cytokine production in follicular helper T cells (Tfh). In two different models, anti-PD-1 mAb increased the activation and proliferation of tumor-specific T cells in lymph nodes. Surprisingly, anti-PD-1 mAb did not primarily target CD8+ T cells but instead stimulated IL-4 production by Tfh cells, the major population bound by anti-PD-1 mAb. Blocking IL-4 or inhibiting the Tfh master transcription factor BCL6 abrogated anti-PD-1 mAb activity in lymph nodes while injection of IL-4 complexes was sufficient to recapitulate anti-PD-1 mAb activity. A similar mechanism was observed in a vaccine model. Finally, nivolumab also boosted human Tfh cells in humanized mice. We propose that Tfh cells and IL-4 play a key role in the peripheral activity of anti-PD-1 mAb.
Collapse
Affiliation(s)
- Mathilde Ruggiu
- Institut Pasteur, Université de Paris Cité, INSERM U1223, Paris, France
| | - Marion V. Guérin
- Institut Pasteur, Université de Paris Cité, INSERM U1223, Paris, France
| | - Béatrice Corre
- Institut Pasteur, Université de Paris Cité, INSERM U1223, Paris, France
| | - Margot Bardou
- Institut Pasteur, Université de Paris Cité, INSERM U1223, Paris, France
| | - Ruby Alonso
- Institut Pasteur, Université de Paris Cité, INSERM U1223, Paris, France
| | - Erica Russo
- Institut Pasteur, Université de Paris Cité, INSERM U1223, Paris, France
| | - Zacarias Garcia
- Institut Pasteur, Université de Paris Cité, INSERM U1223, Paris, France
| | - Lea Feldmann
- Institut Pasteur, Université de Paris Cité, INSERM U1223, Paris, France
| | - Fabrice Lemaître
- Institut Pasteur, Université de Paris Cité, INSERM U1223, Paris, France
| | | | | | - Philippe Bousso
- Institut Pasteur, Université de Paris Cité, INSERM U1223, Paris, France
- Vaccine Research Institute, Creteil, France
| |
Collapse
|
283
|
Lai J, Kuang X, Fu Y, Li J. Association between sex and efficacy of immune checkpoint inhibitors: a systematic review and meta-analysis. Immunotherapy 2024; 16:481-495. [PMID: 38420849 DOI: 10.2217/imt-2023-0307] [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: 11/20/2023] [Accepted: 02/04/2024] [Indexed: 03/02/2024] Open
Abstract
Aim: To explore the association between sex and immune checkpoint inhibitors (ICIs). Materials & methods: We assessed the difference in survival outcomes from ICIs between sexes using an interaction test. Results: 108 studies representing 70,243 patients were included. In the first-line setting, the pooled interaction HR was 0.97 (95% CI: 0.91-1.04). In the subsequent-line setting, the pooled interaction HR was 0.85 (95% CI: 0.77-0.95). When ICIs were given as perioperative therapy or as systemic therapy in patients with positive PD-L1 expression, both men and women obtained equal survival benefits. Conclusion: Both sex, line of therapy, cancer (sub)type and PD-L1 status should be taken into account in the assessment of risk versus benefit when deciding to offer ICIs to patients.
Collapse
Affiliation(s)
- Jianxiong Lai
- Department of General Surgery, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, 621000, China
| | - Xiaohong Kuang
- Department of Hematology, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, 621000, China
| | - Yi Fu
- Department of General Surgery, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, 621000, China
| | - Jian Li
- Department of General Surgery, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, 621000, China
| |
Collapse
|
284
|
Goh LY, Limbachia K, Moonim M, Morley AMS. Primary lacrimal sac melanoma: a case report describing the novel use of fine needle aspiration cytology (FNAC) for diagnosis, together with literature review and immunotherapy treatment update. Orbit 2024; 43:270-279. [PMID: 36069101 DOI: 10.1080/01676830.2022.2119264] [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: 07/24/2022] [Accepted: 08/25/2022] [Indexed: 10/14/2022]
Abstract
Primary lacrimal sac melanoma (PLSM) is exceedingly rare and associated with high morbidity and mortality. Unfortunately, PLSM often presents insidiously resulting in delayed detection and poor prognosis. A 69-year-old Black man was suspected of having a lacrimal sac tumour following presentation with a left sided watery eye, bloody tears, and a lacrimal mass. Due to the patient's implantable pacemaker, defibrillator, and high anticoagulation, an ultrasound-guided FNAC was performed instead of incisional biopsy, revealing a PLSM. Diagnosis was confirmed following complete tumour resection with free flap reconstruction and neck dissection. Unfortunately, disease progression ensued despite further neck dissection and three cycles of both pembrolizumab and iplimumab. This is the first description of FNAC to accurately diagnose PLSM and highlights its use as an accurate, rapid, and minimally invasive technique that may allow an earlier screening diagnosis of lacrimal sac tumours. We also discuss the outcome of immunotherapy in recent similar cases.
Collapse
Affiliation(s)
- Li Yen Goh
- Department of Ophthalmology, Guy's and St Thomas' Hospital NHS Trust, London, UK
| | - Ketan Limbachia
- Department of Ophthalmology, Guy's and St Thomas' Hospital NHS Trust, London, UK
| | - Mufaddal Moonim
- Department of Histopathology, Guy's and St Thomas' Hospital NHS Trust, London, UK
| | - Ana M S Morley
- Department of Ophthalmology, Guy's and St Thomas' Hospital NHS Trust, London, UK
| |
Collapse
|
285
|
Jani Y, Jansen CS, Gerke MB, Bilen MA. Established and emerging biomarkers of immunotherapy in renal cell carcinoma. Immunotherapy 2024; 16:405-426. [PMID: 38264827 PMCID: PMC11913054 DOI: 10.2217/imt-2023-0267] [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: 10/17/2023] [Accepted: 01/08/2024] [Indexed: 01/25/2024] Open
Abstract
Immunotherapies, such as immune checkpoint inhibitors, have heralded impressive progress for patient care in renal cell carcinoma (RCC). Despite this success, some patients' disease fails to respond, and other patients experience significant side effects. Thus, development of biomarkers is needed to ensure that patients can be selected to maximize benefit from immunotherapies. Improving clinicians' ability to predict which patients will respond to immunotherapy and which are most at risk of adverse events - namely through clinical biomarkers - is indispensable for patient safety and therapeutic efficacy. Accordingly, an evolving suite of therapeutic biomarkers continues to be investigated. This review discusses biomarkers for immunotherapy in RCC, highlighting current practices and emerging innovations, aiming to contribute to improved outcomes for patients with RCC.
Collapse
Affiliation(s)
- Yash Jani
- Mercer University, Macon, GA31207, USA
| | - Caroline S Jansen
- Emory University School of Medicine, Atlanta, GA30322, USA
- Winship Cancer Institute of Emory University, Atlanta, GA30322, USA
| | - Margo B Gerke
- Emory University School of Medicine, Atlanta, GA30322, USA
| | - Mehmet Asim Bilen
- Winship Cancer Institute of Emory University, Atlanta, GA30322, USA
- Department of Hematology & Medical Oncology, Emory University School of Medicine, Atlanta, GA30322, USA
| |
Collapse
|
286
|
Euvrard R, Robert M, Mainbourg S, Dalle S, Lega JC. Association between immune-related adverse events and prognosis in patients treated with immune checkpoint inhibitors in melanoma: A surrogacy analysis. Fundam Clin Pharmacol 2024; 38:369-379. [PMID: 38012082 DOI: 10.1111/fcp.12966] [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: 09/28/2022] [Revised: 07/10/2023] [Accepted: 10/26/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICI) represent a breakthrough in oncology in terms of prognosis and safety. They now constitute a cornerstone in the management of metastatic melanoma. However, a new kind of adverse event called immune-related adverse events (irAE) has emerged. These irAE could be conceptually considered as an indicator of the antitumoral immune response, but the association between irAE and prognosis is still a matter of debate. OBJECTIVE The purpose of this study was to investigate the association between the overall survival (OS) and the prevalence of irAE in melanoma. METHODS MEDLINE/PubMed, WebofScience, ClinicalTrials, and WHOTrials databases were searched to identify phase 3 randomized controlled trials (RCT) assessing ICI in melanoma and published up to April 2021. A weighted regression was performed to estimate this association according to standard method of surrogacy analysis. RESULTS A total of 14 RCT including 7646 patients (median age: 59.3 years) with melanoma were included. All types of ICI were represented (ipilimumab, tremelimumab, pembrolizumab, nivolumab, atezolizumab, as well as ipilimumab and nivolumab combination). irAE were frequent but rarely fatal. The combination of ICI caused more irAE than anti-PD1 (or PDL1) and anti-CTLA4 monotherapies. No relationship was found between the occurrence of irAE and OS (beta coefficient 0.078, R2 3%, p = 0.52), nor between cutaneous irAE and OS (beta coefficient 0.080, R2 6%, p = 0.33). CONCLUSION Although limited by the heterogeneity of ICI included in the regression and the low number of included RCT, the present study suggests an absence of association between irAE and prognosis in melanoma.
Collapse
Affiliation(s)
- Romain Euvrard
- Service de Médecine Interne et Pathologie Vasculaire, Hôpital Lyon Sud, Hospices Civils de Lyon, France
| | - Marie Robert
- Service de Médecine Interne et d'immunologie clinique, Université de Lyon 1, Hôpital Édouard Herriot, Hospices Civils de Lyon, Lyon, France
| | - Sabine Mainbourg
- Equipe Évaluation et Modélisations des Effets Thérapeutiques, UMR CRNS 5558, Université Claude Bernard Lyon 1, Lyon, France
- Lyon Immunopathology Federation (LIFe), Hospices Civils de Lyon, France
| | - Stéphane Dalle
- Service de Dermatologie, Hôpital Lyon Sud, Hospices Civils de Lyon, France
- ImmuCare (Immunology Cancer Research), Hospices Civils de Lyon, France
| | - Jean-Christophe Lega
- Equipe Évaluation et Modélisations des Effets Thérapeutiques, UMR CRNS 5558, Université Claude Bernard Lyon 1, Lyon, France
- Lyon Immunopathology Federation (LIFe), Hospices Civils de Lyon, France
- ImmuCare (Immunology Cancer Research), Hospices Civils de Lyon, France
- Service de Rhumatologie, Hôpital Lyon Sud, Hospices Civils de lyon, France
| |
Collapse
|
287
|
Patel MR, Johnson M, Winer I, Arkenau HT, Cook N, Samouëlian V, Aljumaily R, Kitano S, Duffy C, Ge M, Elgadi M, Siu LL. Ezabenlimab (BI 754091), an anti-PD-1 antibody, in patients with advanced solid tumours. Cancer Immunol Immunother 2024; 73:89. [PMID: 38554156 PMCID: PMC10981579 DOI: 10.1007/s00262-024-03654-0] [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: 03/17/2023] [Accepted: 02/09/2024] [Indexed: 04/01/2024]
Abstract
BACKGROUND Ezabenlimab (BI 754091) is a humanised monoclonal antibody targeting programmed cell death protein-1. We report results from open-label, dose-escalation/expansion, Phase I trials that evaluated the safety, maximum tolerated dose (MTD), pharmacokinetics and antitumour activity of ezabenlimab at the recommended Phase II dose in patients with selected advanced solid tumours. STUDY DESIGN Study 1381.1 (NCT02952248) was conducted in Canada, the United Kingdom and the United States. Study 1381.4 (NCT03433898) was conducted in Japan. Study 1381.3 (NCT03780725) was conducted in the Netherlands. The primary endpoints were: number of patients experiencing dose-limiting toxicities (DLTs) in the first cycle (dose escalation parts), number of patients with DLTs during the entire treatment period and objective response (dose expansion part of Study 1381.1). RESULTS Overall, 117 patients received ezabenlimab intravenously every 3 weeks (80 mg, n = 3; 240 mg, n = 111; 400 mg, n = 3). No DLTs were observed and the MTD was not reached. Fifty-eight patients (52.3%) had grade ≥ 3 adverse events, most commonly anaemia (10.8%) and fatigue (2.7%). In 111 assessed patients treated with ezabenlimab 240 mg, disease control rate was 56.8% and objective response rate was 16.2%. Three patients had complete response; at data cut-off (November 2021) one remained in response and was still receiving ongoing treatment (duration of response [DoR]: 906 days). Partial responses occurred across several tumour types; DoR ranged from 67 to 757 days. CONCLUSIONS Ezabenlimab was well tolerated and associated with durable antitumour activity in multiple solid tumours, comparable to other immune checkpoint inhibitors in similar patient populations and treatment settings.
Collapse
Affiliation(s)
- Manish R Patel
- Sarah Cannon Research Institute, 250 25th Ave N, Nashville, TN, 37203, USA.
- Florida Cancer Specialists, 600 N Cattlemen Rd, Suite #200, Sarasota, FL, 34232, USA.
| | - Melissa Johnson
- Sarah Cannon Research Institute, 250 25th Ave N, Nashville, TN, 37203, USA
- Tennessee Oncology, Nashville, TN, USA
| | - Ira Winer
- Wayne State School of Medicine, Karmanos Cancer Institute, Detroit, MI, USA
| | - Hendrik-Tobias Arkenau
- Sarah Cannon Research Institute, Cancer Institute, University College London, London, UK
| | - Natalie Cook
- The Christie NHS Foundation Trust and the University of Manchester, Manchester, UK
| | | | - Raid Aljumaily
- Sarah Cannon Research Institute, 250 25th Ave N, Nashville, TN, 37203, USA
- Stephenson Cancer Center of the University of Oklahoma and Sarah Cannon Research Institute, Oklahoma City, OK, USA
| | - Shigehisa Kitano
- Japanese Foundation for Cancer Research, Tokyo, Japan
- National Cancer Center Hospital, Tokyo, Japan
| | - Christine Duffy
- Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, USA
| | - Miaomiao Ge
- Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, USA
| | - Mabrouk Elgadi
- Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, USA
| | - Lillian L Siu
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| |
Collapse
|
288
|
Dillen A, Bui I, Jung M, Agioti S, Zaravinos A, Bonavida B. Regulation of PD-L1 Expression by YY1 in Cancer: Therapeutic Efficacy of Targeting YY1. Cancers (Basel) 2024; 16:1237. [PMID: 38539569 PMCID: PMC10968822 DOI: 10.3390/cancers16061237] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/23/2024] [Accepted: 03/11/2024] [Indexed: 05/14/2025] Open
Abstract
During the last decade, we have witnessed several milestones in the treatment of various resistant cancers including immunotherapeutic strategies that have proven to be superior to conventional treatment options, such as chemotherapy and radiation. This approach utilizes the host's immune response, which is triggered by cancer cells expressing tumor-associated antigens or neoantigens. The responsive immune cytotoxic CD8+ T cells specifically target and kill tumor cells, leading to tumor regression and prolongation of survival in some cancers; however, some cancers may exhibit resistance due to the inactivation of anti-tumor CD8+ T cells. One mechanism by which the anti-tumor CD8+ T cells become dysfunctional is through the activation of the inhibitory receptor programmed death-1 (PD-1) by the corresponding tumor cells (or other cells in the tumor microenvironment (TME)) that express the programmed death ligand-1 (PD-L1). Hence, blocking the PD-1/PD-L1 interaction via specific monoclonal antibodies (mAbs) restores the CD8+ T cells' functions, leading to tumor regression. Accordingly, the Food and Drug Administration (FDA) has approved several checkpoint antibodies which act as immune checkpoint inhibitors. Their clinical use in various resistant cancers, such as metastatic melanoma and non-small-cell lung cancer (NSCLC), has shown significant clinical responses. We have investigated an alternative approach to prevent the expression of PD-L1 on tumor cells, through targeting the oncogenic transcription factor Yin Yang 1 (YY1), a known factor overexpressed in many cancers. We report the regulation of PD-L1 by YY1 at the transcriptional, post-transcriptional, and post-translational levels, resulting in the restoration of CD8+ T cells' anti-tumor functions. We have performed bioinformatic analyses to further explore the relationship between both YY1 and PD-L1 in cancer and to corroborate these findings. In addition to its regulation of PD-L1, YY1 has several other anti-cancer activities, such as the regulation of proliferation and cell viability, invasion, epithelial-mesenchymal transition (EMT), metastasis, and chemo-immuno-resistance. Thus, targeting YY1 will have a multitude of anti-tumor activities resulting in a significant obliteration of cancer oncogenic activities. Various strategies are proposed to selectively target YY1 in human cancers and present a promising novel therapeutic approach for treating unresponsive cancer phenotypes. These findings underscore the distinct regulatory roles of YY1 and PD-L1 (CD274) in cancer progression and therapeutic response.
Collapse
Affiliation(s)
- Ana Dillen
- Department of Microbiology, Immunology & Molecular Genetics, Jonsson Comprehensive Cancer, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; (A.D.); (I.B.)
| | - Indy Bui
- Department of Microbiology, Immunology & Molecular Genetics, Jonsson Comprehensive Cancer, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; (A.D.); (I.B.)
| | - Megan Jung
- Department of Microbiology, Immunology & Molecular Genetics, Jonsson Comprehensive Cancer, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; (A.D.); (I.B.)
| | - Stephanie Agioti
- Cancer Genetics, Genomic and Systems Biology Group, Basic and Translational Cancer Research Center (BTCRC), 1516 Nicosia, Cyprus (A.Z.)
| | - Apostolos Zaravinos
- Cancer Genetics, Genomic and Systems Biology Group, Basic and Translational Cancer Research Center (BTCRC), 1516 Nicosia, Cyprus (A.Z.)
- Department of Life Sciences, School of Sciences, European University Cyprus, 2404 Nicosia, Cyprus
| | - Benjamin Bonavida
- Department of Microbiology, Immunology & Molecular Genetics, Jonsson Comprehensive Cancer, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; (A.D.); (I.B.)
| |
Collapse
|
289
|
Larkin J, Marais R, Porta N, Gonzalez de Castro D, Parsons L, Messiou C, Stamp G, Thompson L, Edmonds K, Sarker S, Banerji J, Lorigan P, Evans TRJ, Corrie P, Marshall E, Middleton MR, Nathan P, Nicholson S, Ottensmeier C, Plummer R, Bliss J, Valpione S, Turajlic S. Nilotinib in KIT-driven advanced melanoma: Results from the phase II single-arm NICAM trial. Cell Rep Med 2024; 5:101435. [PMID: 38417447 PMCID: PMC10982988 DOI: 10.1016/j.xcrm.2024.101435] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 12/01/2023] [Accepted: 01/26/2024] [Indexed: 03/01/2024]
Abstract
Mucosal (MM) and acral melanomas (AM) are rare melanoma subtypes of unmet clinical need; 15%-20% harbor KIT mutations potentially targeted by small-molecule inhibitors, but none yet approved in melanoma. This multicenter, single-arm Phase II trial (NICAM) investigates nilotinib safety and activity in KIT mutated metastatic MM and AM. KIT mutations are identified in 39/219 screened patients (18%); of 29/39 treated, 26 are evaluable for primary analysis. Six patients were alive and progression free at 6 months (local radiology review, 25%); 5/26 (19%) had objective response at 12 weeks; median OS was 7.7 months; ddPCR assay correctly identifies KIT alterations in circulating tumor DNA (ctDNA) in 16/17 patients. Nilotinib is active in KIT-mutant AM and MM, comparable to other KIT inhibitors, with demonstrable activity in nonhotspot KIT mutations, supporting broadening of KIT evaluation in AM and MM. Our results endorse further investigations of nilotinib for the treatment of KIT-mutated melanoma. This clinical trial was registered with ISRCTN (ISRCTN39058880) and EudraCT (2009-012945-49).
Collapse
Affiliation(s)
- James Larkin
- Skin and Renal Units, The Royal Marsden Hospital NHS Foundation Trust, London, UK; Melanoma and Kidney Cancer Team, The Institute of Cancer Research, London, UK
| | - Richard Marais
- Cancer Research UK Manchester Institute, The University of Manchester, Manchester, UK
| | - Nuria Porta
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, UK
| | - David Gonzalez de Castro
- Molecular Diagnostics, The Institute of Cancer Research and Royal Marsden NHS Foundation Trust, London, UK
| | - Lisa Parsons
- University of Edinburgh, Edinburgh, UK; PDD - Thermo Fisher Scientific, Bend, Oregon, USA
| | - Christina Messiou
- Department of Radiology, The Royal Marsden Hospital NHS Foundation Trust, London, UK
| | - Gordon Stamp
- Department of Histopathology, The Royal Marsden Hospital NHS Foundation Trust, London, UK
| | - Lisa Thompson
- Centre for Molecular Pathology, The Royal Marsden Hospital NHS Foundation Trust, London, UK
| | - Kim Edmonds
- Skin and Renal Units, The Royal Marsden Hospital NHS Foundation Trust, London, UK
| | - Sarah Sarker
- Skin and Renal Units, The Royal Marsden Hospital NHS Foundation Trust, London, UK
| | - Jane Banerji
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, UK
| | - Paul Lorigan
- Division of Cancer Sciences, Unviersity of Manchester, Manchester, UK; The Christie NHS Foundation Trust, Manchester, UK
| | | | - Pippa Corrie
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Ernest Marshall
- The Clatterbridge Cancer Centre NHS Foundation Trust, Liverpool, UK
| | | | - Paul Nathan
- Mount Vernon Cancer Centre, East & North Herts NHS Trust, Northwood, UK
| | - Steve Nicholson
- University Hospitals of Leicester NHS Foundation Trust, Leicester, UK
| | | | - Ruth Plummer
- Newcastle University and Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle, UK
| | - Judith Bliss
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, UK
| | - Sara Valpione
- Cancer Research UK Manchester Institute, The University of Manchester, Manchester, UK; The Christie NHS Foundation Trust, Manchester, UK.
| | - Samra Turajlic
- Skin and Renal Units, The Royal Marsden Hospital NHS Foundation Trust, London, UK; Melanoma and Kidney Cancer Team, The Institute of Cancer Research, London, UK; Cancer Dynamics Laboratory, The Francis Crick Institute, London, UK.
| |
Collapse
|
290
|
Vardarli I, Tan S, Brandenburg T, Weidemann F, Görges R, Herrmann K, Führer D. Risk and Incidence of Endocrine Immune-Related Adverse Effects Under Checkpoint Inhibitor Mono- or Combination Therapy in Solid Tumors: A Meta-Analysis of Randomized Controlled Trials. J Clin Endocrinol Metab 2024; 109:1132-1144. [PMID: 37967245 DOI: 10.1210/clinem/dgad670] [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: 06/04/2023] [Revised: 11/10/2023] [Accepted: 11/13/2023] [Indexed: 11/17/2023]
Abstract
CONTEXT Few meta-analyses on incidence of endocrine immune-related adverse effects (eirAEs) have been published and many trials have been published since. OBJECTIVE We performed a comprehensive meta-analysis with updated literature to assess risk and incidence of eirAEs of any grade and grade 3 to 5 by immune checkpoint inhibitor (ICI) monotherapy or combination therapy in solid tumors. METHODS An electronic search using PubMed/Medline, Embase, and the Cochrane Library was performed. Randomized controlled studies (RCTs) assessing eirAEs under ICI monotherapy or ICI combination therapy were selected. Stata software (v17) was used for statistical analyses and risk of bias was evaluated using Review Manager version 5.3. RESULTS A total of 69 RCTs with 80 independent reports, involving 42 886 patients, were included in the study. Meta-analysis revealed the following pooled estimates for risk ratio and incidence, respectively: for any grade hypothyroidism 7.81 (95% CI, 5.68-10.74, P < .0001) and 7.64% (95% CI, 6.23-9.17, P < .0001); significantly increased also for hyperthyroidism, hypophysitis/hypopituitarism, and adrenal insufficiency; and for insulin-dependent diabetes mellitus 1.52 (95% CI, 1.07-2.18, P = .02), and 0.087% (95% CI, 0.019-0.189, P = .0006), respectively. Meta-regression showed that combination of ICIs (nivolumab plus ipilimumab; durvalumab plus tremelimumab) is an independent risk factor for any grade hypophysitis/hypopituitarism, and that ICI agent is an independent factor of risk for adrenal insufficiency, but that cancer type is not an independent risk factor for eirAEs. CONCLUSION We showed that risk, independent from cancer type, and incidence of eirAEs are substantially increased with ICI therapy. Combination of ICIs increases risk for eirAEs, especially for hypophysitis/hypopituitarism.
Collapse
Affiliation(s)
- Irfan Vardarli
- 5th Medical Department, Division of Endocrinology and Diabetes, Medical Faculty Mannheim, Heidelberg University, Mannheim 68167, Germany
- Department of Medicine I, Klinikum Vest, Knappschaftskrankenhaus Recklinghausen, Academic Teaching Hospital, Ruhr-University Bochum, Recklinghausen 45657, Germany
| | - Susanne Tan
- Department of Endocrinology, Diabetes and Metabolism, Clinical Chemistry-Division of Laboratory Research; Endocrine Tumor Center at WTZ/Comprehensive Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen 45147, Germany
| | - Tim Brandenburg
- Department of Endocrinology, Diabetes and Metabolism, Clinical Chemistry-Division of Laboratory Research; Endocrine Tumor Center at WTZ/Comprehensive Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen 45147, Germany
| | - Frank Weidemann
- Department of Medicine I, Klinikum Vest, Knappschaftskrankenhaus Recklinghausen, Academic Teaching Hospital, Ruhr-University Bochum, Recklinghausen 45657, Germany
| | - Rainer Görges
- Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Essen 45147, Germany
| | - Ken Herrmann
- Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Essen 45147, Germany
| | - Dagmar Führer
- Department of Endocrinology, Diabetes and Metabolism, Clinical Chemistry-Division of Laboratory Research; Endocrine Tumor Center at WTZ/Comprehensive Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen 45147, Germany
| |
Collapse
|
291
|
Wong P, Wisneski AD, Tsai KK, Chang TT, Hirose K, Nakakura EK, Daud AI, Maker AV, Corvera CU. Metastatic melanoma to small bowel: metastasectomy is supported in the era of immunotherapy and checkpoint inhibitors. World J Surg Oncol 2024; 22:77. [PMID: 38468341 PMCID: PMC10926580 DOI: 10.1186/s12957-024-03335-3] [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: 11/28/2023] [Accepted: 02/14/2024] [Indexed: 03/13/2024] Open
Abstract
BACKGROUND Metastatic melanoma to the small bowel is an aggressive disease often accompanied by obstruction, abdominal pain, and gastrointestinal bleeding. With advancements in melanoma treatment, the role for metastasectomy continues to evolve. Inclusion of novel immunotherapeutic agents, such as checkpoint inhibitors, into standard treatment regimens presents potential survival benefits for patients receiving metastasectomy. CASE PRESENTATION We report an institutional experience of 15 patients (12 male, 3 female) between 2014-2022 that underwent small bowel metastasectomy for metastatic melanoma and received perioperative systemic treatment. Median age of patients was 64 years (range: 35-83 years). No patients died within 30 days of their surgery, and the median hospital length of stay was 5 days. Median overall survival in these patients was 30.1 months (range: 2-115 months). Five patients died from disease (67 days, 252 days, 426 days, 572 days, 692 days postoperatively), one patient died of non-disease related causes (1312 days postoperatively), six patients are alive with disease, and three remain disease free. CONCLUSIONS This case series presents an updated perspective of the utility of metastasectomy for small bowel metastasis in the age of novel immunotherapeutic agents as standard systemic treatment. Small bowel metastasectomy for advanced melanoma performed in conjunction with perioperative systemic therapy is safe and appears to promote long-term survival and enhanced quality of life.
Collapse
Affiliation(s)
- Paul Wong
- Division of Surgical Oncology, Department of Surgery, University of California, San Francisco, 505 Parnassus Ave, S549, Box 1932, San Francisco, California, 94143-1932, USA
| | - Andrew D Wisneski
- Division of Surgical Oncology, Department of Surgery, University of California, San Francisco, 505 Parnassus Ave, S549, Box 1932, San Francisco, California, 94143-1932, USA
| | - Katy K Tsai
- Division of Hematology and Oncology, Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Tammy T Chang
- Division of Surgical Oncology, Department of Surgery, University of California, San Francisco, 505 Parnassus Ave, S549, Box 1932, San Francisco, California, 94143-1932, USA
| | - Kenzo Hirose
- Division of Surgical Oncology, Department of Surgery, University of California, San Francisco, 505 Parnassus Ave, S549, Box 1932, San Francisco, California, 94143-1932, USA
| | - Eric K Nakakura
- Division of Surgical Oncology, Department of Surgery, University of California, San Francisco, 505 Parnassus Ave, S549, Box 1932, San Francisco, California, 94143-1932, USA
| | - Adil I Daud
- Division of Hematology and Oncology, Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Ajay V Maker
- Division of Surgical Oncology, Department of Surgery, University of California, San Francisco, 505 Parnassus Ave, S549, Box 1932, San Francisco, California, 94143-1932, USA
| | - Carlos U Corvera
- Division of Surgical Oncology, Department of Surgery, University of California, San Francisco, 505 Parnassus Ave, S549, Box 1932, San Francisco, California, 94143-1932, USA.
| |
Collapse
|
292
|
Pasello G, Fabricio ASC, Del Bianco P, Salizzato V, Favaretto A, Piccin L, Zustovich F, Fabozzi A, De Rossi C, Pigozzo J, De Nuzzo M, Cappelletto E, Bonanno L, Palleschi D, De Salvo GL, Guarneri V, Gion M, Chiarion-Sileni V. Sex-related differences in serum biomarker levels predict the activity and efficacy of immune checkpoint inhibitors in advanced melanoma and non-small cell lung cancer patients. J Transl Med 2024; 22:242. [PMID: 38443899 PMCID: PMC10916307 DOI: 10.1186/s12967-024-04920-6] [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: 11/30/2023] [Accepted: 01/20/2024] [Indexed: 03/07/2024] Open
Abstract
BACKGROUND Immune Checkpoint Inhibitors (ICIs) lead to durable response and a significant increase in long-term survival in patients with advanced malignant melanoma (MM) and Non-Small Cell Lung Cancer (NSCLC). The identification of serum cytokines that can predict their activity and efficacy, and their sex interaction, could improve treatment personalization. METHODS In this prospective study, we enrolled immunotherapy-naïve patients affected by advanced MM and NSCLC treated with ICIs. The primary endpoint was to dissect the potential sex correlations between serum cytokines (IL-1β, IL-2, IL-4, IL-5, IL-6, IL-8, IL-10, GM-CSF, MCP-1, TNF-ɑ, IP-10, VEGF, sPD-L1) and the objective response rate (ORR). Secondly, we analyzed biomarker changes during treatment related to ORR, disease control rate (DCR), progression free survival (PFS) and overall survival (OS). Blood samples, collected at baseline and during treatment until disease progression (PD) or up to 2 years, were analyzed using Luminex xMAP or ELLA technologies. RESULTS Serum samples from 161 patients (98 males/63 females; 92 MM/69 NSCLC) were analyzed for treatment response. At baseline, IL-6 was significantly lower in females (F) versus males (M); lower levels of IL-4 in F and of IL-6 in both sexes significantly correlated with a better ORR, while higher IL-4 and TNF-ɑ values were predictive of a lower ORR in F versus M. One hundred and sixty-five patients were evaluable for survival analysis: at multiple Cox regression, an increased risk of PD was observed in F with higher baseline values of IL-4, sPD-L1 and IL-10, while higher IL-6 was a negative predictor in males. In males, higher levels of GM-CSF predict a longer survival, whereas higher IL-1β predicts a shorter survival. Regardless of sex, high baseline IL-8 values were associated with an increased risk of both PD and death, and high IL-6 levels only with shorter OS. CONCLUSIONS Serum IL-1β, IL-4, IL-6, IL-10, GM-CSF, TNF-ɑ, and sPD-L1 had a significant sex-related predictive impact on ORR, PFS and OS in melanoma and NSCLC patients treated with ICIs. These results will potentially pave the way for new ICI combinations, designed according to baseline and early changes of these cytokines and stratified by sex.
Collapse
Affiliation(s)
- Giulia Pasello
- Medical Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy.
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy.
| | - Aline S C Fabricio
- Medical Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Paola Del Bianco
- Clinical Research Unit, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | | | - Adolfo Favaretto
- Medical Oncology Unit, Ca' Foncello Hospital, AULSS 2, Treviso, Italy
| | - Luisa Piccin
- Medical Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | | | - Alessio Fabozzi
- Medical Oncology 3, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | | | - Jacopo Pigozzo
- Medical Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Mattia De Nuzzo
- Medical Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - Elia Cappelletto
- Regional Center for Biomarkers, Department of Clinical Pathology, AULSS3 Serenissima, Venice, Italy
| | - Laura Bonanno
- Medical Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Dario Palleschi
- Medical Oncology Unit, Ca' Foncello Hospital, AULSS 2, Treviso, Italy
| | - Gian Luca De Salvo
- Clinical Research Unit, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Valentina Guarneri
- Medical Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - Massimo Gion
- Regional Center for Biomarkers, Department of Clinical Pathology, AULSS3 Serenissima, Venice, Italy
| | | |
Collapse
|
293
|
Kim HJ, Kim YH. Molecular Frontiers in Melanoma: Pathogenesis, Diagnosis, and Therapeutic Advances. Int J Mol Sci 2024; 25:2984. [PMID: 38474231 DOI: 10.3390/ijms25052984] [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: 01/02/2024] [Revised: 03/01/2024] [Accepted: 03/02/2024] [Indexed: 03/14/2024] Open
Abstract
Melanoma, a highly aggressive skin cancer, is characterized by rapid progression and high mortality. Recent advances in molecular pathogenesis have shed light on genetic and epigenetic changes that drive melanoma development. This review provides an overview of these developments, focusing on molecular mechanisms in melanoma genesis. It highlights how mutations, particularly in the BRAF, NRAS, c-KIT, and GNAQ/GNA11 genes, affect critical signaling pathways. The evolution of diagnostic techniques, such as genomics, transcriptomics, liquid biopsies, and molecular biomarkers for early detection and prognosis, is also discussed. The therapeutic landscape has transformed with targeted therapies and immunotherapies, improving patient outcomes. This paper examines the efficacy, challenges, and prospects of these treatments, including recent clinical trials and emerging strategies. The potential of novel treatment strategies, including neoantigen vaccines, adoptive cell transfer, microbiome interactions, and nanoparticle-based combination therapy, is explored. These advances emphasize the challenges of therapy resistance and the importance of personalized medicine. This review underlines the necessity for evidence-based therapy selection in managing the increasing global incidence of melanoma.
Collapse
Affiliation(s)
- Hyun Jee Kim
- Department of Dermatology, International St. Mary's Hospital, College of Medicine, Catholic Kwandong University, Incheon 22711, Republic of Korea
| | - Yeong Ho Kim
- Department of Dermatology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
294
|
Kopecký J, Pásek M, Lakomý R, Melichar B, Mrazová I, Kubeček O, Arenbergerová M, Lemstrová R, Švancarová A, Tretera V, Hlodáková A, Žváčková K. The outcome in patients with BRAF-mutated metastatic melanoma treated with anti-programmed death receptor-1 monotherapy or targeted therapy in the real-world setting. Cancer Med 2024; 13:e6982. [PMID: 38491825 PMCID: PMC10943370 DOI: 10.1002/cam4.6982] [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: 07/08/2023] [Revised: 10/24/2023] [Accepted: 01/15/2024] [Indexed: 03/18/2024] Open
Abstract
BACKGROUND Immunotherapy and targeted therapy are currently two alternative backbones in the therapy of BRAF-mutated malignant melanoma. However, predictive biomarkers that would help with treatment selection are lacking. METHODS This retrospective study investigated outcomes of anti-programmed death receptor-1 monotherapy and targeted therapy in the first-line setting in patients with metastatic BRAF-mutated melanoma, focusing on clinical and laboratory parameters associated with treatment outcome. RESULTS Data from 174 patients were analysed. The median progression-free survival (PFS) was 17.0 months (95% CI; 8-39) and 12.5 months (95% CI; 9-14.2) for immunotherapy and targeted therapy, respectively. The 3-year PFS rate was 39% for immunotherapy and 25% for targeted therapy. The objective response rate was 72% and 51% for targeted therapy and immunotherapy. The median overall (OS) survival for immunotherapy has not been reached and was 23.6 months (95% CI; 16.1-38.2) for targeted therapy, with a 3-year survival rate of 63% and 40%, respectively. In a univariate analysis, age < 70 years, a higher number of metastatic sites, elevated serum LDH and a neutrophil-lymphocyte ratio above the cut-off value were associated with inferior PFS regardless of the therapy received, but only serum LDH level and the presence of lung metastases remained significant predictors of PFS in a multivariate analysis. CONCLUSIONS Present real-world data document the high effectiveness of immunotherapy and targeted therapy. Although targeted therapy had higher response rates, immunotherapy improved PFS and OS. While the prognostic value of LDH was confirmed, the potential use of blood cell count-derived parameters to predict outcomes needs further investigation.
Collapse
Affiliation(s)
- Jindřich Kopecký
- Department of Clinical Radiotherapy and OncologyUniversity Hospital in Hradec KraloveHradec KraloveCzech Republic
| | - Marek Pásek
- Department of Dermatovenereology, Third Faculty of MedicineCharles University and Kralovske Vinohrady University HospitalPragueCzech Republic
| | - Radek Lakomý
- Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute and Faculty of MedicineMasaryk UniversityBrnoCzech Republic
| | - Bohuslav Melichar
- Department of Oncology, Faculty of Medicine and DentistryPalacký University and University HospitalOlomoucCzech Republic
| | - Ivona Mrazová
- Department of OncologyCounty HospitalČeské BudějoviceCzech Republic
| | - Ondřej Kubeček
- Department of Clinical Radiotherapy and OncologyUniversity Hospital in Hradec KraloveHradec KraloveCzech Republic
| | - Monika Arenbergerová
- Department of Dermatovenereology, Third Faculty of MedicineCharles University and Kralovske Vinohrady University HospitalPragueCzech Republic
| | - Radmila Lemstrová
- Department of Oncology, Faculty of Medicine and DentistryPalacký University and University HospitalOlomoucCzech Republic
| | - Alžběta Švancarová
- Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute and Faculty of MedicineMasaryk UniversityBrnoCzech Republic
| | - Vojtěch Tretera
- Department of Dermatovenereology, Third Faculty of MedicineCharles University and Kralovske Vinohrady University HospitalPragueCzech Republic
| | - Alžběta Hlodáková
- Department of Clinical Radiotherapy and OncologyUniversity Hospital in Hradec KraloveHradec KraloveCzech Republic
| | - Kamila Žváčková
- Department of Oncology, Faculty of Medicine and DentistryPalacký University and University HospitalOlomoucCzech Republic
| |
Collapse
|
295
|
Di Giacomo AM, Schenker M, Medioni J, Mandziuk S, Majem M, Gravis G, Cornfeld M, Ranganathan S, Lou S, Csoszi T. A phase II study of retifanlimab, a humanized anti-PD-1 monoclonal antibody, in patients with solid tumors (POD1UM-203). ESMO Open 2024; 9:102387. [PMID: 38401247 PMCID: PMC10982862 DOI: 10.1016/j.esmoop.2024.102387] [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: 06/09/2023] [Revised: 01/25/2024] [Accepted: 01/26/2024] [Indexed: 02/26/2024] Open
Abstract
BACKGROUND POD1UM-203, an open-label, multicenter, phase II study, evaluated retifanlimab, a humanized monoclonal antibody targeting programmed cell death protein-1 (PD-1) in patients with selected solid tumors where immune checkpoint inhibitor therapies have previously shown efficacy. PATIENTS AND METHODS Eligible patients (≥18 years) had measurable disease and included unresectable or metastatic melanoma, treatment-naive metastatic non-small-cell lung cancer (NSCLC) with high programmed death-ligand 1 (PD-L1) expression (tumor proportion score ≥50%), cisplatin-ineligible locally advanced/metastatic urothelial carcinoma (UC) with PD-L1 expression (combined positive score ≥10%), or treatment-naive locally advanced/metastatic clear-cell renal cell carcinoma (RCC). Retifanlimab 500 mg was administered intravenously every 4 weeks as a 30-min infusion. The primary endpoint was investigator-assessed overall response rate. RESULTS Overall, 121 patients (35 melanoma, 23 NSCLC, 29 UC, 34 RCC) were enrolled and treated. The overall response rate [95% confidence interval (CI)] was 40.0% (23.9-57.9) in the melanoma cohort, 34.8% (16.4-57.3) in the NSCLC cohort, 37.9% (20.7-57.7) in the UC cohort, and 23.5% (10.7-41.2) in the RCC cohort. Median duration of response was 11.5 months (95% CI 2.2-not reached) in the UC cohort, and was not reached in the other cohorts. Retifanlimab safety was consistent with previous experience for PD-(L)1 inhibitors. CONCLUSIONS Retifanlimab demonstrated durable antitumor activity in patients with melanoma, NSCLC, UC, or RCC. The efficacy and safety of retifanlimab were as expected for a PD-(L)1 inhibitor. These data support further study of retifanlimab in solid tumors.
Collapse
Affiliation(s)
- A M Di Giacomo
- University of Siena, Siena, Italy; Center for Immuno-Oncology, University Hospital of Siena, Siena, Italy
| | - M Schenker
- Centrul de Oncologie Sf. Nectarie, Oncologie Medicala, Craiova, Romania
| | - J Medioni
- Centre of Early Clinical Trials in Cancer, Hôpital Européen Georges-Pompidou, Université Paris Cité, Paris, France
| | - S Mandziuk
- Department of Clinical Oncology and Chemotherapy, Medical University of Lublin, Lublin, Poland
| | - M Majem
- Medical Oncology Department, Hospital de Sant Pau, Barcelona, Spain
| | - G Gravis
- Department of Medical Oncology, Institut Paoli-Calmettes, Aix-Marseille Université, CRCM, Marseille, France
| | | | | | - S Lou
- Incyte Corporation, Wilmington, USA
| | - T Csoszi
- Hetényi Géza Kórház Onkológiai Központ, Szolnok, Hungary.
| |
Collapse
|
296
|
Dnyanmote AS, Jadhav S, Vasava K, Immadi S. Anorectal Melanoma: A Rare Cause of Large Bowel Obstruction. Cureus 2024; 16:e56128. [PMID: 38618462 PMCID: PMC11015112 DOI: 10.7759/cureus.56128] [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] [Accepted: 03/13/2024] [Indexed: 04/16/2024] Open
Abstract
Anorectal melanoma is a rare and aggressive malignancy with a challenging diagnosis and management. We present the case of a 69-year-old male with a history of chronic constipation and recent weight loss, who presented with symptoms suggestive of anorectal pathology. Despite initial diagnostic challenges, including an unsuccessful colonoscopy due to inadequate bowel preparation, the subsequent radiographic evaluation revealed a large bowel obstruction secondary to a protruding anorectal mass. Abdominal X-ray demonstrated significant colonic dilation while contrast-enhanced CT scan revealed a large hypodense mass protruding from the anal canal. Surgical intervention led to the excision of the mass, and histopathological examination confirmed malignant melanoma. Immunohistochemistry markers, including HMB 45, Melan A, and S100, supported the diagnosis. This case underscores the importance of considering anorectal melanoma in patients presenting with atypical anorectal symptoms, despite its rarity. Early recognition and intervention, supported by appropriate imaging modalities, are critical for optimizing patient outcomes in such cases.
Collapse
Affiliation(s)
- Anuradha S Dnyanmote
- Surgery, Dr. D. Y. Patil Medical College, Hospital and Research Centre, Pune, IND
| | - Suhasini Jadhav
- General Surgery, Dr. D. Y. Patil Medical College, Hospital and Research Centre, Pune, IND
| | - Kinjal Vasava
- General Surgery, Dr. D. Y. Patil Medical College, Hospital and Research Centre, Pune, IND
| | - Saikumar Immadi
- General Surgery, Dr. D. Y. Patil Medical College, Hospital and Research Centre, Pune, IND
| |
Collapse
|
297
|
Shah V, Panchal V, Shah A, Vyas B, Agrawal S, Bharadwaj S. Immune checkpoint inhibitors in metastatic melanoma therapy (Review). MEDICINE INTERNATIONAL 2024; 4:13. [PMID: 38410760 PMCID: PMC10895472 DOI: 10.3892/mi.2024.137] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 01/26/2024] [Indexed: 02/28/2024]
Abstract
An increase in the incidence of melanoma has been observed in recent decades, which poses a significant challenge due to its poor prognosis in the advanced and metastatic stages. Previously, chemotherapy and high doses of interleukin-2 were available treatments for melanoma; however, they offered limited survival benefits and were associated with severe toxicities. The treatment of metastatic melanoma has been transformed by new developments in immunotherapy. Immune checkpoint inhibitors (ICIs), monoclonal antibodies that target cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4), programmed cell death protein 1 (PD-1) and its ligand, PDL-1, have emerged as promising therapeutic options. Commonly used ICIs, such as ipilimumab, nivolumab and pembrolizumab, have been found to be associated with an improved median overall survival, recurrence-free survival and response rates compared to traditional chemotherapies. Combination therapies involving different types of ICIs, such as anti-PD1 with anti-CTLA-4, have further enhanced the overall survival and response rates by targeting various phases of T-cell activation. Additionally, the development of novel biomarkers has facilitated the assessment of responses to ICI therapy, with tissue and serum-based prognostic and predictive biomarkers now available. The increased response observed with ICIs also provides potential for immune-related adverse effects on various organ systems. Further research is required to evaluate the efficacy and safety of various combinations of ICIs, while ongoing clinical trials explore the potential of newer ICIs. Concerns regarding the development of resistance to ICIs also warrant attention. The present review summarizes and discusses the advent of ICIs with a marked significant breakthrough in the treatment of metastatic melanoma, providing improved outcomes compared to traditional therapies.
Collapse
Affiliation(s)
- Vedant Shah
- Department of Medicine, Smt. N.H.L. Municipal Medical College and Sardar Vallabhbhai Patel Institute of Medical Sciences and Research (SVPISMR), Ahmedabad, Gujarat 380058, India
| | - Viraj Panchal
- Department of Medicine, Smt. N.H.L. Municipal Medical College and Sardar Vallabhbhai Patel Institute of Medical Sciences and Research (SVPISMR), Ahmedabad, Gujarat 380058, India
| | - Abhi Shah
- Department of Medicine, Smt. N.H.L. Municipal Medical College and Sardar Vallabhbhai Patel Institute of Medical Sciences and Research (SVPISMR), Ahmedabad, Gujarat 380058, India
| | - Bhavya Vyas
- Department of Medicine, Smt. N.H.L. Municipal Medical College and Sardar Vallabhbhai Patel Institute of Medical Sciences and Research (SVPISMR), Ahmedabad, Gujarat 380058, India
| | - Siddharth Agrawal
- Department of Medicine, Smt. N.H.L. Municipal Medical College and Sardar Vallabhbhai Patel Institute of Medical Sciences and Research (SVPISMR), Ahmedabad, Gujarat 380058, India
| | - Sanket Bharadwaj
- Department of Medicine, Smt. N.H.L. Municipal Medical College and Sardar Vallabhbhai Patel Institute of Medical Sciences and Research (SVPISMR), Ahmedabad, Gujarat 380058, India
| |
Collapse
|
298
|
Zhang X, Xu Q, Zhang Y. Remarkable response to pazopanib plus vivolumab in a patient with pericardial synovial sarcoma carrying a novel genotype BRCA2 c.968dupT: A case report. Thorac Cancer 2024; 15:667-671. [PMID: 38323364 DOI: 10.1111/1759-7714.15237] [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: 11/30/2023] [Accepted: 01/20/2024] [Indexed: 02/08/2024] Open
Abstract
Pericardial synovial sarcomas (PSS) have a low incidence rate and are highly invasive with a dismal prognosis. Standard treatment includes surgery, radiotherapy and chemotherapy but with limited response. Here, we report the case of a 15-year-old nonsmoking youngster diagnosed with PSS who developed disease relapsed from surgery after 1 month. Next-generation sequencing (NGS) using baseline tissue was performed, and BRCA2 c.968dupT was detected. Then pazopanib (a multitargeted inhibitor) plus nivolumab (an immune checkpoint inhibitor) was administered, with a partial response and progression-free survival of 14 months. BRCA2 c.968dupT has not previously been reported in PSS and its response to targeted combination immunotherapy are not well characterized. Here, we report the efficacy of pazopanib combined with nivolumab in a PSS patient harboring BRCA2 c.968dupT and also provide the clinical evidence of the utility of NGS in exploring actionable mutations for solid tumor. Combination therapy based on immunotherapy may be a potential treatment choice for PSS harboring BRCA2 mutation.
Collapse
Affiliation(s)
- Xing Zhang
- Graduate Collaborative Training Base of Hunan Cancer Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital, Department of Medical Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Changsha, Hunan, China
| | - Qinqin Xu
- Department of Medical Oncology, Qinghai Provincial People's Hospital, Xining, Qinghai, China
| | - Yongchang Zhang
- Graduate Collaborative Training Base of Hunan Cancer Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital, Department of Medical Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Changsha, Hunan, China
| |
Collapse
|
299
|
Uchi H. Optimal strategy in managing advanced melanoma. J Dermatol 2024; 51:324-334. [PMID: 38087810 PMCID: PMC11483965 DOI: 10.1111/1346-8138.17068] [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: 09/30/2023] [Revised: 11/01/2023] [Accepted: 11/20/2023] [Indexed: 03/05/2024]
Abstract
The advent of immune checkpoint inhibitors and combination therapy with BRAF inhibitors and MEK inhibitors has dramatically improved the prognosis of advanced melanoma. However, since acral melanoma and mucosal melanoma, which are rare in Western countries but are major subtypes of melanoma in East Asia, including Japan, have a low frequency of BRAF mutations, there are currently no treatment options other than immune checkpoint inhibitors in most such cases. Furthermore, owing to a lower tumor mutation burden, immune checkpoint inhibitors are less effective in acral and mucosal melanoma than in cutaneous melanoma. The aim of this review was to summarize the current status and future prospects for the treatment of advanced melanoma, comparing cutaneous melanoma, acral melanoma, and mucosal melanoma.
Collapse
Affiliation(s)
- Hiroshi Uchi
- Department of Dermato‐OncologyNational Hospital Organization Kyushu Cancer CenterFukuokaJapan
| |
Collapse
|
300
|
Namikawa K, Nakano E, Ogata D, Yamazaki N. Long-term survival with systemic therapy in the last decade: Can melanoma be cured? J Dermatol 2024; 51:343-352. [PMID: 38358050 PMCID: PMC11484129 DOI: 10.1111/1346-8138.17147] [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: 01/14/2024] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 02/16/2024]
Abstract
Immune checkpoint inhibitors have been shown to prolong survival of patients with several types of cancer, and the finding was first established in melanoma. Previously, systemic therapy for advanced melanoma aimed only at tumor control and palliation of symptoms. However, in recent years, some patients who received systemic therapy have achieved a complete response and survived without continuous treatment for more than several years. This review discusses the long-term survival rates achieved with currently used systemic therapies and their future perspectives. Long-term survival is currently most likely to be achieved with the use of the standard-dose combination of nivolumab plus ipilimumab, however, this regimen is associated with a high frequency of serious or persistent immune-related adverse events. Several new anti-PD-1-based combination therapies with a better risk-benefit balance are currently under development. Although the acral and mucosal subtypes tend to be less responsive to immune checkpoint inhibitors, anti-PD-1-based combination therapy should continue to be investigated for these subtypes owing to its potential for better long-term survival. With the development of efficacious immunotherapy and targeted therapy, it is important to determine the optimal duration of systemic therapy to avoid unnecessary health and financial burdens as well as to improve efforts to support long-term cancer survivors. As the goal of systemic therapy shifts from tumor control to long-term survival, in future clinical trials, long-term clinical outcomes should be evaluated to assess the benefits of novel agents.
Collapse
Affiliation(s)
- Kenjiro Namikawa
- Department of Dermatologic OncologyNational Cancer Center HospitalTokyoJapan
| | - Eiji Nakano
- Department of Dermatologic OncologyNational Cancer Center HospitalTokyoJapan
| | - Dai Ogata
- Department of Dermatologic OncologyNational Cancer Center HospitalTokyoJapan
| | - Naoya Yamazaki
- Department of Dermatologic OncologyNational Cancer Center HospitalTokyoJapan
| |
Collapse
|