1
|
Gu Y, Ly A, Rodriguez S, Zhang H, Kim J, Mao Z, Sachdeva A, Zomorodian N, Pellegrini M, Li G, Liu S, Drakaki A, Rettig MB, Chin AI. PD-1 blockade plus cisplatin-based chemotherapy in patients with small cell/neuroendocrine bladder and prostate cancers. Cell Rep Med 2024:101824. [PMID: 39536751 DOI: 10.1016/j.xcrm.2024.101824] [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: 04/30/2024] [Revised: 08/14/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024]
Abstract
Small cell neuroendocrine cancers share biologic similarities across tissue types, including transient response to platinum-based chemotherapy with rapid progression of disease. We report a phase 1b study of pembrolizumab in combination with platinum-based chemotherapy in 15 patients with stage III-IV small cell bladder (cohort 1) or small cell/neuroendocrine prostate cancers (cohort 2). Overall response rate (ORR) is 43% with two-year overall survival (OS) rate of 86% (95% confidence interval [CI]: 0.63, 1.00) for cohort 1 and 57% (95% CI: 0.30, 1.00) for cohort 2. Treatment is tolerated well with grade 3 or higher adverse events occurring in 40% of patients with no deaths or treatment cessation secondary to toxicity. Single-cell and T cell receptor sequencing of serial peripheral blood samples reveals clonal expansion of diverse T cell repertoire correlating with progression-free survival. Our results demonstrate promising efficacy and safety of this treatment combination and support future investigation of this biomarker. This study was registered at ClinicalTrials.gov (NCT03582475).
Collapse
Affiliation(s)
- Yiqian Gu
- Department of Molecular, Cellular and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ann Ly
- Department of Urology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Sara Rodriguez
- Department of Urology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Hanwei Zhang
- Department of Urology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jiyoon Kim
- Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA, USA
| | - Zhiyuan Mao
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ankush Sachdeva
- Department of Urology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Nazy Zomorodian
- Department of Urology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Matteo Pellegrini
- Department of Molecular, Cellular and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA
| | - Gang Li
- Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Sandy Liu
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Alexandra Drakaki
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Matthew B Rettig
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; The VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Arnold I Chin
- Department of Urology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
2
|
Provencio M, Nadal E, Insa A, García Campelo R, Casal J, Dómine M, Massuti B, Majem M, Rodríguez-Abreu D, Martínez-Martí A, de Castro J, Gómez de Antonio D, Macia I, Figueroa S, Fernández Vago L, Calvo V, Palmero R, Sierra-Rodero B, Martínez-Toledo C, Molina-Alejandre M, Serna-Blasco R, Romero A, Cruz-Bermúdez A. Perioperative chemotherapy and nivolumab in non-small-cell lung cancer (NADIM): 5-year clinical outcomes from a multicentre, single-arm, phase 2 trial. Lancet Oncol 2024; 25:1453-1464. [PMID: 39419061 PMCID: PMC11519011 DOI: 10.1016/s1470-2045(24)00498-4] [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: 07/17/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND Perioperative immunotherapy improves short-term outcomes in resectable non-small-cell lung cancer (NSCLC). We now report 5-year survival from the NADIM trial to assess its long-term benefit. METHODS NADIM was a multicentre, single-arm, phase 2 trial conducted across 18 hospitals in Spain. Patients were aged 18 years or older, had an Eastern Cooperative Oncology Group performance status of 0 or 1, and had histologically or cytologically confirmed, treatment-naive, resectable stage IIIA NSCLC (American Joint Committee on Cancer, 7th edition criteria). The neoadjuvant treatment consisted of three cycles of intravenous paclitaxel (200 mg/m2) and carboplatin (area under the curve 6 mg/mL per min) with nivolumab (360 mg). After surgery, 1 year of adjuvant treatment with intravenous nivolumab monotherapy was administered (240 mg every 2 weeks for 4 months, followed by 480 mg every 4 weeks for 8 months). The primary endpoint was 24-month progression-free survival, with 5-year progression-free survival and overall survival as secondary endpoints, assessed in the intention-to-treat population (ie, all patients who received neoadjuvant treatment). Toxicity profile was also assessed as a secondary endpoint. This trial is registered at ClinicalTrials.gov (NCT03081689) and is complete; this is the final report of the trial. FINDINGS Between April 26, 2017, and Aug 25, 2018, 51 patients were assessed for eligibility, of whom 46 comprised the intention-to-treat population (34 [74%] male and 12 [26%] female, median age 63 years [IQR 58-70]). Follow-up was concluded at 60 months (data cutoff July 11, 2023; median follow-up 60·0 months [IQR 60·0-60·0]). 5-year progression-free survival in the intention-to-treat population was 65·0% (95% CI 49·4-76·9), and overall survival was 69·3% (53·7-80·6). Disease progression occurred in 11 (24%) patients; 14 (30%) patients died, including nine (20%) from disease relapse and five (11%) from non-tumour-related causes. Treatment-related adverse events (TRAEs) of grade 3 or worse occurred in 14 (30%) of 46 patients during neoadjuvant treatment and in seven (19%) of 37 during adjuvant treatment. The most common grade 3 or worse TRAEs were increased lipase and febrile neutropenia (three [7%] each) during neoadjuvant treatment, and elevated serum lipase (four [7%]) and elevated serum amylase (three [8%]) during adjuvant treatment. Serious TRAEs included elevated serum lipase and neutropenia (one [2%] each) during neoadjuvant treatment, and elevated serum lipase (one [3%]) during adjuvant treatment. No treatment-related surgery delays, deaths, or unexpected long-term toxicities were reported. INTERPRETATION Perioperative chemoimmunotherapy showed a promising long-term benefit with no concerning safety data, reinforcing its use in resectable stage IIIA NSCLC. FUNDING Bristol-Myers Squibb, Spanish Ministry of Science, Instituto de Salud Carlos III, European Union.
Collapse
Affiliation(s)
- Mariano Provencio
- Servicio de Oncología Médica, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDIPHISA), Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain.
| | - Ernest Nadal
- Institut Català d'Oncologia, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Amelia Insa
- Fundación INCLIVA, Hospital Clínico Universitario de Valencia, Valencia, Spain
| | | | | | - Manuel Dómine
- Hospital Universitario Fundación Jiménez Díaz, Madrid, Spain
| | - Bartomeu Massuti
- Hospital General Dr. Balmis de Alicante, ISABIAL, Alicante, Spain
| | | | | | - Alex Martínez-Martí
- Hospital Universitario e Instituto de Oncología Vall d'Hebron (VHIO), Barcelona, Spain
| | | | - David Gómez de Antonio
- Servicio de Cirugía Torácica, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDIPHISA), Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Iván Macia
- Institut Català d'Oncologia, L'Hospitalet de Llobregat, Barcelona, Spain; Department of Thoracic Surgery, Hospital Universitari de Bellvitge, Barcelona, Spain; Department of Pathology and Experimental Therapeutics, Universitat de Barcelona, Barcelona, Spain; Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Santiago Figueroa
- Fundación INCLIVA, Hospital Clínico Universitario de Valencia, Valencia, Spain
| | | | - Virginia Calvo
- Servicio de Oncología Médica, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDIPHISA), Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Ramón Palmero
- Institut Català d'Oncologia, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Belén Sierra-Rodero
- Servicio de Oncología Médica, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDIPHISA), Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Cristina Martínez-Toledo
- Servicio de Oncología Médica, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDIPHISA), Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Marta Molina-Alejandre
- Servicio de Oncología Médica, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDIPHISA), Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Roberto Serna-Blasco
- Servicio de Oncología Médica, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDIPHISA), Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Atocha Romero
- Servicio de Oncología Médica, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDIPHISA), Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Alberto Cruz-Bermúdez
- Servicio de Oncología Médica, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDIPHISA), Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| |
Collapse
|
3
|
Molina-Alejandre M, Perea F, Calvo V, Martinez-Toledo C, Nadal E, Sierra-Rodero B, Casarrubios M, Casal-Rubio J, Martinez-Martí A, Insa A, Massuti B, Viteri S, Barneto Aranda I, Rodriguez-Abreu D, de Castro J, Martínez JM, Cobo M, Wistuba II, Parra ER, Martín-López J, Megías D, Muñoz-Viana R, Garrido F, Aptsiauri N, Ruiz-Cabello F, Provencio M, Cruz-Bermúdez A. Perioperative chemoimmunotherapy induces strong immune responses and long-term survival in patients with HLA class I-deficient non-small cell lung cancer. J Immunother Cancer 2024; 12:e009762. [PMID: 39428126 PMCID: PMC11492944 DOI: 10.1136/jitc-2024-009762] [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: 05/27/2024] [Accepted: 09/26/2024] [Indexed: 10/22/2024] Open
Abstract
BACKGROUND Loss of human leukocyte antigen (HLA) class I expression and loss of heterozygosity (LOH) are common events implicated in the primary resistance of non-small cell lung cancer (NSCLC) to immunotherapy. However, there is no data on perioperative chemoimmunotherapy (ChIO) efficacy or response mechanisms in the context of HLA class I defects. METHODS Baseline HLA class I tumor status (HLA-deficient (HLA-DEF) or HLA-proficient (HLA-PRO)) was determined by DNA LOH combined with immunohistochemistry for protein levels in tissue of 24 patients with NSCLC treated with perioperative nivolumab plus chemotherapy from NADIM trial (NCT03081689). We integrated HLA tumor status with molecular data (programmed death-ligand 1 (PD-L1), TMB, TCR repertoire, TILs populations, bulk RNA-seq, and spatial transcriptomics (ST)) and clinical outcomes (pathological response and survival data) to study the activity of perioperative ChIO considering HLA class I defects. RESULTS HLA-DEF tumors comprised 41.7% of analyzed tumors and showed a desert-like microenvironment at baseline, with lower PD-L1 levels and reduced immune infiltrate. However, perioperative ChIO induced similar complete pathological response (CPR) rates in both HLA-DEF and PRO tumors (50% and 60% respectively, p=0.670), as well as 3-year survival rates: Progression-free survival (PFS) and overall survival (OS) of 70% (95% CI 32.9% to 89.2%) for HLA-DEF, and PFS 71.4% (95% CI 40.6% to 88.2%) and OS 92.9% (95% CI 59.1% to 99.0%) for HLA-PRO (log-rank PFS p=0.909, OS p=0.137). Proof-of-concept ST analysis of a CPR HLA-DEF tumor after ChIO showed a strong immune response with tertiary lymphoid structures (TLS), CD4+T cells with HLA class II colocalization, and activated CD8+T cells. CONCLUSIONS Our findings highlight the activity of perioperative ChIO, and the potential role of TLS and T-cell immune response, in NSCLC HLA-DEF tumors.
Collapse
Affiliation(s)
- Marta Molina-Alejandre
- Departament of Medical Oncology, Hospital Universitario Puerta de Hierro Majadahonda, Majadahonda, Comunidad de Madrid, Spain
| | - Francisco Perea
- Departamento de Bioquímica, Biología Molecular e Inmunología III. Instituto de Investigación Biosanitaria de Granada (Ibs. GRANADA), Universidad de Granada Facultad de Medicina, Granada, Andalucía, Spain
| | - Virginia Calvo
- Departament of Medical Oncology, Hospital Universitario Puerta de Hierro Majadahonda, Majadahonda, Comunidad de Madrid, Spain
| | - Cristina Martinez-Toledo
- Departament of Medical Oncology, Hospital Universitario Puerta de Hierro Majadahonda, Majadahonda, Comunidad de Madrid, Spain
| | - Ernest Nadal
- Catalan Institute of Oncology. Oncobell Program. IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Belén Sierra-Rodero
- Departament of Medical Oncology, Hospital Universitario Puerta de Hierro Majadahonda, Majadahonda, Comunidad de Madrid, Spain
| | - Marta Casarrubios
- Departament of Medical Oncology, Hospital Universitario Puerta de Hierro Majadahonda, Majadahonda, Comunidad de Madrid, Spain
| | | | - Alex Martinez-Martí
- Departament of Medical Oncology, Hospital Universitari Vall d'Hebron, Barcelona, Catalunya, Spain
| | - Amelia Insa
- Fundación INCLIVA, Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - Bartomeu Massuti
- Hospital General Universitario Dr. Balmis de Alicante, Alicante, Spain
| | - Santiago Viteri
- Hospital Universitario Quiron Dexeus, Grupo Quironsalud, Barcelona, Catalunya, Spain
| | | | | | | | | | - Manuel Cobo
- Medical Oncology Intercenter Unit. IBIMA, Virgen de la Victoria University Hospital Pharmacy Clinic Management Unit, Malaga, Andalucía, Spain
| | - Ignacio I Wistuba
- The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Edwin R Parra
- The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Javier Martín-López
- Pathological Anatomy, Hospital Universitario Puerta de Hierro Majadahonda, Majadahonda, Spain
| | - Diego Megías
- Unidad de Microscopía Óptica Avanzada, Instituto de Salud Carlos III, Majadahonda, Spain
| | - Rafael Muñoz-Viana
- Unidad de Bioinformática, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana, Majadahonda, Spain
| | - Federico Garrido
- Departamento de Bioquímica, Biología Molecular e Inmunología III. Instituto de Investigación Biosanitaria de Granada (Ibs. GRANADA), Universidad de Granada Facultad de Medicina, Granada, Andalucía, Spain
| | - Natalia Aptsiauri
- Departamento de Bioquímica, Biología Molecular e Inmunología III. Instituto de Investigación Biosanitaria de Granada (Ibs. GRANADA), Universidad de Granada Facultad de Medicina, Granada, Andalucía, Spain
| | - Francisco Ruiz-Cabello
- Departamento de Bioquímica, Biología Molecular e Inmunología III. Instituto de Investigación Biosanitaria de Granada (Ibs. GRANADA), Universidad de Granada Facultad de Medicina, Granada, Andalucía, Spain
| | - Mariano Provencio
- Departament of Medical Oncology, Hospital Universitario Puerta de Hierro Majadahonda, Majadahonda, Comunidad de Madrid, Spain
| | - Alberto Cruz-Bermúdez
- Departament of Medical Oncology, Hospital Universitario Puerta de Hierro Majadahonda, Majadahonda, Comunidad de Madrid, Spain
| |
Collapse
|
4
|
Wang D, Liu S, Fu J, Zhang P, Zheng S, Qiu B, Liu H, Ye Y, Guo J, Zhou Y, Jiang H, Yin S, He H, Xie C, Liu H. Correlation of K trans derived from dynamic contrast-enhanced MRI with treatment response and survival in locally advanced NSCLC patients undergoing induction immunochemotherapy and concurrent chemoradiotherapy. J Immunother Cancer 2024; 12:e008574. [PMID: 38910009 PMCID: PMC11328668 DOI: 10.1136/jitc-2023-008574] [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: 05/30/2024] [Indexed: 06/25/2024] Open
Abstract
PURPOSE This study aimed to investigate the prognostic significance of pretreatment dynamic contrast-enhanced (DCE)-MRI parameters concerning tumor response following induction immunochemotherapy and survival outcomes in patients with locally advanced non-small cell lung cancer (NSCLC) who underwent immunotherapy-based multimodal treatments. MATERIAL AND METHODS Unresectable stage III NSCLC patients treated by induction immunochemotherapy, concurrent chemoradiotherapy (CCRT) with or without consolidative immunotherapy from two prospective clinical trials were screened. Using the two-compartment Extend Tofts model, the parameters including Ktrans, Kep, Ve, and Vp were calculated from DCE-MRI data. The apparent diffusion coefficient was calculated from diffusion-weighted-MRI data. The receiver operating characteristic (ROC) curve and the area under the curve (AUC) were used to assess the predictive performance of MRI parameters. The Cox regression model was used for univariate and multivariate analysis. RESULTS 111 unresectable stage III NSCLC patients were enrolled. Patients received two cycles of induction immunochemotherapy and CCRT, with or without consolidative immunotherapy. With the median follow-up of 22.3 months, the median progression-free survival (PFS) and overall survival (OS) were 16.3 and 23.8 months. The multivariate analysis suggested that Eastern Cooperative Oncology Group score, TNM stage and the response to induction immunochemotherapy were significantly related to both PFS and OS. After induction immunochemotherapy, 67 patients (59.8%) achieved complete response or partial response and 44 patients (40.2%) had stable disease or progressive disease. The Ktrans of primary lung tumor before induction immunochemotherapy yielded the best performance in predicting the treatment response, with an AUC of 0.800. Patients were categorized into two groups: high-Ktrans group (n=67, Ktrans>164.3×10-3/min) and low-Ktrans group (n=44, Ktrans≤164.3×10-3/min) based on the ROC analysis. The high-Ktrans group had a significantly higher objective response rate than the low-Ktrans group (85.1% (57/67) vs 22.7% (10/44), p<0.001). The high-Ktrans group also presented better PFS (median: 21.1 vs 11.3 months, p=0.002) and OS (median: 34.3 vs 15.6 months, p=0.035) than the low-Ktrans group. CONCLUSIONS Pretreatment Ktrans value emerged as a significant predictor of the early response to induction immunochemotherapy and survival outcomes in unresectable stage III NSCLC patients who underwent immunotherapy-based multimodal treatments. Elevated Ktrans values correlated positively with enhanced treatment response, leading to extended PFS and OS durations.
Collapse
Affiliation(s)
- DaQuan Wang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China
| | - SongRan Liu
- Department of Pathology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China
| | - Jia Fu
- Department of Pathology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China
| | - PengXin Zhang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China
| | - ShiYang Zheng
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China
| | - Bo Qiu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China
| | - Hui Liu
- United Imaging Healthcare, ShangHai, China
| | - YongQuan Ye
- United Imaging of Healthcare America, Houston, Texas, USA
| | - JinYu Guo
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China
| | - Yin Zhou
- SuZhou TongDiao Company, Suzhou, China
| | | | - ShaoHan Yin
- Department of Radiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China
| | - HaoQiang He
- Department of Radiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China
| | - ChuanMiao Xie
- Department of Radiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China
| | - Hui Liu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
5
|
Kaira K, Ichiki Y, Imai H, Kawasaki T, Hashimoto K, Kuji I, Kagamu H. Potential predictors of the pathologic response after neoadjuvant chemoimmunotherapy in resectable non-small cell lung cancer: a narrative review. Transl Lung Cancer Res 2024; 13:1137-1149. [PMID: 38854945 PMCID: PMC11157365 DOI: 10.21037/tlcr-24-142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 03/27/2024] [Indexed: 06/11/2024]
Abstract
Background and Objective Neoadjuvant chemoimmunotherapy (NACI) is the standard of care for patients with resectable non-small cell lung cancer (NSCLC). Although the pathological complete response (pCR) after NACI reportedly exceeds 20%, an optimal predictor of pCR is yet to be established. This review aims to examine the possible predictors of pCR after NACI. Methods We identified research article published between 2018 and 2022 in English by the PubMed database. Fifty research studies were considered as relevant article, and were examined to edit information for this narrative review. Key Content and Findings Recently, several studies have explored potential biomarkers for the pathological response after NACI. For example, 18F-fluorodeoxyglucose positron emission tomography (18F-FDG-PET) imaging, tumor microenvironment (TME), genetic alternation such as circulating tumor DNA (ctDNA), and clinical markers such as neutrophil-to-lymphocyte ratio (NLR) and smoking signature were assessed in patients with resectable NSCLC to predict the pathological response after NACI. Based on the PET response criteria, the complete metabolic response (CMR) achieved a positive predictive value (PPV) of 71.4% for predicting pCR, and the decreasing rate of post-therapy maximum standardized uptake value (SUVmax) after NACI substantially correlated with the major pathological response (MPR). TME, as a significant marker for MPR in tumor specimens, was identified as an increase in CD8+ T cells and decrease in CD3+ T cells or Foxp3 T cells. Considering blood samples, TME comprised an increase in CD4+PD-1+ cells or natural killer cells and a decrease in CD3+CD56+CTLA4+ cells, total T cells, Th cells, myeloid-derived suppressor cells (MDSCs), or regulatory T cells. Although low pretreatment levels of ctDNA and undetectable ctDNA levels after NACI were markedly associated with survival, the relationship between ctDNA levels and pCR remains elusive. Moreover, the patients with a high baseline NLR had a low incidence of pCR. Heavy smoking (>40 pack-years) was favorable for predicting pathological response. Conclusions A reduced rate of 18F-FDG uptake post-NACI and TME-related surface markers on lymphocytes could be optimal predictors for pCR. However, the role of these pCR predictors for NACI remains poorly validated, warranting further investigations. This review focuses on predictive biomarkers for pathological response after NACI in patients with resectable NSCLC.
Collapse
Affiliation(s)
- Kyoichi Kaira
- Department of Respiratory Medicine, International Medical Center, Saitama Medical University, Hidaka, Saitama, Japan
| | - Yoshinobu Ichiki
- Department of General Thoracic Surgery, International Medical Center, Saitama Medical University, Hidaka, Saitama, Japan
| | - Hisao Imai
- Department of Respiratory Medicine, International Medical Center, Saitama Medical University, Hidaka, Saitama, Japan
| | - Tomonori Kawasaki
- Department of Diagnostic Pathology, International Medical Center, Saitama Medical University, Hidaka, Saitama, Japan
| | - Kosuke Hashimoto
- Department of Respiratory Medicine, International Medical Center, Saitama Medical University, Hidaka, Saitama, Japan
| | - Ichiei Kuji
- Department of Nuclear Medicine, International Medical Center, Saitama Medical University, Hidaka, Saitama, Japan
| | - Hiroshi Kagamu
- Department of Respiratory Medicine, International Medical Center, Saitama Medical University, Hidaka, Saitama, Japan
| |
Collapse
|
6
|
Abed A, Beasley AB, Reid AL, Law N, Calapre L, Millward M, Lo J, Gray ES. Circulating pre-treatment T-cell receptor repertoire as a predictive biomarker in advanced or metastatic non-small-cell lung cancer patients treated with pembrolizumab alone or in combination with chemotherapy. ESMO Open 2023; 8:102066. [PMID: 37995426 PMCID: PMC10774950 DOI: 10.1016/j.esmoop.2023.102066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 10/17/2023] [Accepted: 10/18/2023] [Indexed: 11/25/2023] Open
Abstract
BACKGROUND The circulating T-cell receptor (TCR) repertoire is a dynamic representation of overall immune responses in an individual. MATERIALS AND METHODS We prospectively collected baseline blood from patients treated with first-line pembrolizumab monotherapy or in combination with chemotherapy. TCR repertoire metrics were correlated with clinical benefit rate (CBR), progression-free survival (PFS), overall survival (OS) and immune-related adverse events (irAEs). We built a logistic regression classifier by fitting all four TCR-β repertoire metrics to the immune checkpoint inhibitor (ICI) CBR data. In the subsequent receiver operating characteristic (ROC) analysis of the resulting logistic regression model probabilities, the best cut-off value was selected to maximise sensitivity to predict CBR to ICI. RESULTS We observed an association between reduced number of unique clones and CBR among patients treated with pembrolizumab monotherapy (cohort 1) [risk ratio = 2.86, 95% confidence interval (CI) 1.04-8.73, P = 0.039]. For patients treated with pembrolizumab plus chemotherapy (cohort 2), increased number of unique clones [hazard ratio (HR) = 2.96, 95% CI 1.28-6.88, P = 0.012] and Shannon diversity (HR = 2.73, 95% CI 1.08-6.87, P = 0.033), and reduced evenness (HR = 0.43, 95% CI 0.21-0.90, P = 0.025) and convergence (HR = 0.41, 95% CI 0.19-0.90, P = 0.027) were associated with improved PFS, while only an increased number of unique clones (HR = 4.62, 95% CI 1.52-14.02, P = 0.007) were associated with improved OS. Logistic regression models combining the TCR repertoire metrics improved the prediction of CBR (cohorts 1 and 2) and were strongly associated with PFS (cohort 1, HR = 0.38, 95% CI 0.19-0.78, P = 0.009) and OS (cohort 2, HR = 0.20, 95% CI 0.05-0.76, P < 0.0001). Reduced TCR conversion was associated with increased frequency of irAEs needing systemic steroid treatment. CONCLUSION Combined pre-treatment circulating TCR metrics might serve as a predictive biomarker for clinical outcomes among patients with advanced non-small-cell lung cancer treated with pembrolizumab alone or in combination with chemotherapy.
Collapse
Affiliation(s)
- A Abed
- Centre for Precision Health, Edith Cowan University, Joondalup; School of Medical and Health Sciences, Edith Cowan University, Joondalup; School of Medicine, University of Western Australia, Crawley.
| | - A B Beasley
- Centre for Precision Health, Edith Cowan University, Joondalup; School of Medical and Health Sciences, Edith Cowan University, Joondalup
| | - A L Reid
- Centre for Precision Health, Edith Cowan University, Joondalup; School of Medical and Health Sciences, Edith Cowan University, Joondalup
| | - N Law
- Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands
| | - L Calapre
- Centre for Precision Health, Edith Cowan University, Joondalup; School of Medical and Health Sciences, Edith Cowan University, Joondalup
| | - M Millward
- School of Medicine, University of Western Australia, Crawley
| | - J Lo
- School of Science, Edith Cowan University, Joondalup, Australia
| | - E S Gray
- Centre for Precision Health, Edith Cowan University, Joondalup; School of Medical and Health Sciences, Edith Cowan University, Joondalup.
| |
Collapse
|
7
|
Takada K, Takamori S, Brunetti L, Crucitti P, Cortellini A. Impact of Neoadjuvant Immune Checkpoint Inhibitors on Surgery and Perioperative Complications in Patients With Non-small-cell Lung Cancer: A Systematic Review. Clin Lung Cancer 2023; 24:581-590.e5. [PMID: 37741717 DOI: 10.1016/j.cllc.2023.08.017] [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] [Received: 04/03/2023] [Revised: 08/11/2023] [Accepted: 08/31/2023] [Indexed: 09/25/2023]
Abstract
Several clinical trials are currently underway to evaluate immune checkpoint inhibitors (ICIs) as neoadjuvant treatment for patients with early-stage non-small-cell lung cancer (NSCLC), and their use in clinical practice is expected to increase in the future. Therefore, a proper assessment of surgical outcomes and perioperative complications after neoadjuvant ICIs is essential to establish recommendations and guidelines. We performed a systematic literature review in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analysis guidelines (PRISMA), searching the PubMed and Scopus databases from the January 1, 2017, to the July 27, 2023, to identify potentially relevant published trials of neoadjuvant ICIs in patients with reseactable NSCLC with available information on surgical outcomes and perioperative complications. A total of 18 studies were included in the review. The rates of surgery cancellation ranged from 0% to 45.8%. Importantly, adverse events (AEs) were the least reported underlying cause, while disease progression caused from 0% to 75% of cancellations. Surgery delays ranged from 0% to 31.3% with AEs as the most frequently reported underlying cause. However, 6 out of 13 trials (46.2%) reported no surgery delays. Conversion rates from minimally invasive to open chest surgery were available for 7 trials and ranged from 0% to 53.8%. Thirty-day mortality rates ranged from 0% to 5.4%, with 11 out of 16 trials reporting 0%. A few reports described perioperative complications in detail. Considering the limited evidence available, we can preliminarily confirm that preoperative ICIs are safe and well tolerated even from the surgical perspective. Additional details on intraoperative findings from prospective controlled trials are needed to establish and disseminate guidelines and recommendations for thoracic surgeons.
Collapse
Affiliation(s)
- Kazuki Takada
- Department of Surgery, Saiseikai Fukuoka General Hospital, Fukuoka, Japan
| | - Shinkichi Takamori
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Leonardo Brunetti
- Medical Oncology Department, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Pierfilippo Crucitti
- Thoracic Surgery Department, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo 200, 00128, Rome, Italy
| | - Alessio Cortellini
- Medical Oncology Department, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy; Department of Surgery and Cancer, Hammersmith Hospital Campus, Imperial College London, London, UK.
| |
Collapse
|
8
|
Sanber K, Rosner S, Forde PM, Marrone KA. Neoadjuvant Immunotherapy for Non-Small Cell Lung Cancer. BioDrugs 2023; 37:775-791. [PMID: 37603233 DOI: 10.1007/s40259-023-00614-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2023] [Indexed: 08/22/2023]
Abstract
Immune checkpoint blockade (ICB) has improved outcomes for patients with advanced non-small cell lung carcinoma (NSCLC). Building off of this, it has been hypothesized that the utilization of ICB early during the disease course may be advantageous, particularly in the neoadjuvant setting prior to definitive surgical resection. Preclinical studies have suggested that a more potent immune response may be induced by neoadjuvant ICB in the presence of a higher antigen burden and intact tumor draining lymph nodes. Recent clinical trials evaluating neoadjuvant ICB with or without chemotherapy combinations in patients with resectable NSCLC led to improved pathological responses and longer event-free survival when neoadjuvant ICB was added to chemotherapy. Surgical outcomes were also supportive of this approach, with encouraging rates of pathological downstaging. Additionally, the availability of pre-treatment biopsy samples and post-treatment surgical resection tissues facilitates the conducting of correlative studies that continue to improve our understanding of the mechanisms of response and resistance to ICB. As long-term survival outcomes from ongoing clinical trials are awaited, several important questions require further investigation, including the optimal duration of neoadjuvant therapy, the clinical endpoints most predictive of long-term outcomes, and translational studies that should be investigated in future trial designs. Additionally, the optimal clinical management of patients with residual disease at the time of surgical resection and those who experience recurrence remains to be determined. In this review, we will (1) discuss the rationale behind neoadjuvant ICB-based therapy in NSCLC, (2) summarize the clinical data available thus far, and (3) highlight unanswered questions that need to be addressed in future studies to maximize the clinical benefits of this approach.
Collapse
Affiliation(s)
- Khaled Sanber
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Johns Hopkins University School of Medicine, 301 Mason Lord Drive, Suite 4500, Baltimore, MD, 21224, USA
| | - Samuel Rosner
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Johns Hopkins University School of Medicine, 301 Mason Lord Drive, Suite 4500, Baltimore, MD, 21224, USA
| | - Patrick M Forde
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Johns Hopkins University School of Medicine, 301 Mason Lord Drive, Suite 4500, Baltimore, MD, 21224, USA
| | - Kristen A Marrone
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Johns Hopkins University School of Medicine, 301 Mason Lord Drive, Suite 4500, Baltimore, MD, 21224, USA.
| |
Collapse
|
9
|
Chen Y, Yan B, Zhang R, Zhao G, You J. Neoadjuvant immunochemotherapy with pembrolizumab plus chemotherapy in resectable non-small cell lung cancer. Heliyon 2023; 9:e19818. [PMID: 37809935 PMCID: PMC10559199 DOI: 10.1016/j.heliyon.2023.e19818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 08/31/2023] [Accepted: 09/01/2023] [Indexed: 10/10/2023] Open
Abstract
Background Neoadjuvant immunotherapy, the focus of current research and treatment modality for long-term survival, has become one of the main options in supporting primary treatment interventions in early NSCLC. Methods This was a retrospective analysis of patients with locally resectable NSCLC who received the neoadjuvant drug pembrolizumab combined with chemotherapy and underwent surgical resection. Pathological responses, PFS and OS in the whole sample and subgroups were analyzed. Results Of the 61 patients included in this retrospective analysis, 31 (50.82%) achieved a pCR, and 38 (62.30%) obtained an MPR. Patients with a pCR had significantly higher OS than the non-pCR group (HR = 0.093, P = 0.0227); patients with an MPR also had significantly elevated OS compared with the non-MPR group (HR = 0.05357, P = 0.0169). Patients with lymph node metastasis after surgery had significantly reduced OS (HR = 0.01607, p = 0.0004) and PFS (HR = 0.08757, p = 0.0004) than those without lymph node metastasis. There was no significant difference in OS and PFS between squamous cell carcinomas (SCC) group and adenocarcinomas (AD) group. No significant differences in OS and PFS were found between patients administered 2 and 3 cycles of neoadjuvant therapy before surgery, between those administered ≤5 and > 5 cycles of adjuvant therapy post-surgery, and between patients with TPS <50% and ≥50% (all P > 0.05). Conclusion Neoadjuvant immunochemotherapy with pembrolizumab plus chemotherapy in non-small cell lung cancer is safe and tolerable. Both pCR and MPR were closely associated with OS and PFS, reflecting a good response of tumor tissues to drug therapy. Lymph node metastasis after surgery was a poor prognostic factor, reducing OS and PFS.
Collapse
Affiliation(s)
- Yulong Chen
- Department of Lung Cancer, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Bo Yan
- Department of Radiotherapy, National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Ran Zhang
- Tianjin Cancer Hospital Airport Hospital National Clinical Research Center for Cancer,Tianjin 300060, China
| | - Gang Zhao
- Department of Pathology, National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Jian You
- Department of Lung Cancer, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| |
Collapse
|
10
|
Conroy MR, Dennehy C, Forde PM. Neoadjuvant immune checkpoint inhibitor therapy in resectable non-small cell lung cancer. Lung Cancer 2023; 183:107314. [PMID: 37541935 DOI: 10.1016/j.lungcan.2023.107314] [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: 01/31/2023] [Revised: 07/20/2023] [Accepted: 07/21/2023] [Indexed: 08/06/2023]
Abstract
Only a minority of lung cancers are resectable at diagnosis, and many of these will eventually relapse. Adjuvant chemotherapy in this setting has a modest survival advantage, and there is significant need for new approaches to improve cure rates. Checkpoint inhibitor immunotherapy has transformed the prognosis for advanced lung cancer, and is increasingly being used in the neoadjuvant setting alone, or in combination with cytotoxic chemotherapy. While this has demonstrated convincing improvements in event-free survival and pathologic response, questions remain over optimal duration of therapy, predictive and prognostic biomarkers, response assessment and combination with other modalities. In addition, these results must be considered in the context of recent positive studies of adjuvant immunotherapy. Here, we summarise preclinical context and clinical trials in this space, discuss areas of controversy and pitfalls, and consider future challenges.
Collapse
Affiliation(s)
- Michael R Conroy
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, United States
| | - Colum Dennehy
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, United States
| | - Patrick M Forde
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, United States.
| |
Collapse
|
11
|
Zhang B, Ren Z, Zhao J, Zhu Y, Huang B, Xiao C, Zhang Y, Deng J, Mao L, Tang L, Lan D, Gao L, Zhang H, Chen G, Luo OJ. Global analysis of HLA-A2 restricted MAGE-A3 tumor antigen epitopes and corresponding TCRs in non-small cell lung cancer. Theranostics 2023; 13:4449-4468. [PMID: 37649599 PMCID: PMC10465222 DOI: 10.7150/thno.84710] [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: 03/27/2023] [Accepted: 07/31/2023] [Indexed: 09/01/2023] Open
Abstract
Background: Advanced non-small cell lung cancer (NSCLC) is the most common type of lung cancer with poor prognosis. Adoptive cell therapy using engineered T-cell receptors (TCRs) targeting cancer-testis antigens, such as Melanoma-associated antigen 3 (MAGE-A3), is a potential approach for the treatment of NSCLC. However, systematic analysis of T cell immune responses to MAGE-A3 antigen and corresponding antigen-specific TCR is still lacking. Methods: In this study, we comprehensively screened HLA-A2 restricted MAGE-A3 tumor epitopes and characterized the corresponding TCRs using in vitro artificial antigen presentation cells (APC) system, single-cell transcriptome and TCR V(D)J sequencing, and machine-learning. Furthermore, the tumor-reactive TCRs with killing potency was screened and verified. Results: We identified the HLA-A2 restricted T cell epitopes from MAGE-A3 that could effectively induce the activation and cytotoxicity of CD8+ T cells using artificial APC in vitro. A cohort of HLA-A2+ NSCLC donors demonstrated that the number of epitope specific CD8+ T cells increased in NSCLC than healthy controls when measured with tetramer derived from the candidate MAGE-A3 epitopes, especially epitope Mp4 (MAGE-A3: 160-169, LVFGIELMEV). Statistical and machine-learning based analyses demonstrated that the MAGE-A3-Mp4 epitope-specific CD8+ T cell clones were mostly in effector and proliferating state. Importantly, T cells artificially expressing the MAGE-A3-Mp4 specific TCRs exhibited strong MAGE-A3+ tumor cell recognition and killing effect. Cross-reactivity risk analysis of the candidates TCRs showed high binding stability to MAGE-A3-Mp4 epitope and low risk of cross-reaction. Conclusions: This work identified candidate TCRs potentially suitable for TCR-T design targeting HLA-A2 restricted MAGE-A3 tumor antigen.
Collapse
Affiliation(s)
- Bei Zhang
- Department of Systems Biomedical Sciences, School of Medicine, Jinan University, Guangzhou, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou, China
| | - Zhiyao Ren
- Guangzhou Geriatric Hospital, Guangzhou, China
- Collaborative Innovation Center for Civil Affairs of Guangzhou, Guangzhou, China
| | - Jianfu Zhao
- Department of Oncology, Research Center of Cancer Diagnosis and Therapy, the First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Yue Zhu
- Department of Systems Biomedical Sciences, School of Medicine, Jinan University, Guangzhou, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou, China
| | - Boya Huang
- Department of Systems Biomedical Sciences, School of Medicine, Jinan University, Guangzhou, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou, China
| | - Chanchan Xiao
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou, China
- Department of Microbiology and Immunology; Institute of Geriatric Immunology; School of Medicine, Jinan University, Guangzhou, China
| | - Yan Zhang
- Department of Oncology, Research Center of Cancer Diagnosis and Therapy, the First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Jieping Deng
- Department of Systems Biomedical Sciences, School of Medicine, Jinan University, Guangzhou, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou, China
| | - Lipeng Mao
- Department of Systems Biomedical Sciences, School of Medicine, Jinan University, Guangzhou, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou, China
| | - Lei Tang
- School of Life Science & Technology, China Pharmaceutical University, Nanjing, China
| | - Dan Lan
- Department of Oncology, Research Center of Cancer Diagnosis and Therapy, the First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Lijuan Gao
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou, China
- Department of Microbiology and Immunology; Institute of Geriatric Immunology; School of Medicine, Jinan University, Guangzhou, China
| | - Hongyi Zhang
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou, China
- Department of Microbiology and Immunology; Institute of Geriatric Immunology; School of Medicine, Jinan University, Guangzhou, China
| | - Guobing Chen
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou, China
- Department of Microbiology and Immunology; Institute of Geriatric Immunology; School of Medicine, Jinan University, Guangzhou, China
| | - Oscar Junhong Luo
- Department of Systems Biomedical Sciences, School of Medicine, Jinan University, Guangzhou, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou, China
| |
Collapse
|
12
|
Yang Y, Liu Z. Neoadjuvant immune checkpoint inhibitor treatment + chemotherapy (vs. chemotherapy alone) for locally advanced non‑small cell lung cancer: A retrospective cohort study. Oncol Lett 2023; 26:292. [PMID: 37274484 PMCID: PMC10236247 DOI: 10.3892/ol.2023.13878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 02/07/2023] [Indexed: 06/06/2023] Open
Abstract
Neoadjuvant immune checkpoint inhibitor (ICI) treatment + chemotherapy has been used for locally advanced non-small cell lung cancer (NSCLC); however, evidence regarding the efficacy of this treatment is insufficient, particularly in Chinese patients. Therefore, the aim of the present study was to evaluate the efficacy and safety of neoadjuvant ICI treatment + chemotherapy compared with neoadjuvant chemotherapy alone for locally advanced NSCLC. For this, 50 patients with locally advanced NSCLC were retrospectively analyzed; of these, 23 patients received pre-operative camrelizumab or sintilimab + chemotherapy (ICI + chemo group) and 27 patients received pre-operative chemotherapy alone (chemo group). The objective response rate (73.9 vs. 44.4%, P=0.035) was superior in the ICI + chemo group compared with the chemo group. Nevertheless, surgical resection rate (100.0 vs. 88.9%, P=0.240), major pathological response (60.9 vs. 41.7%, P=0.188) and complete pathological response (CPR; 30.4 vs. 8.3%, P=0.072) were not significantly different in the ICI + chemo group compared with the chemo group. Following adjustment, ICI + chemo was independently associated with an elevated CPR (P=0.029). Disease-free survival (DFS) was prolonged in the ICI + chemo group compared with the chemo group (1-year DFS, 94.1 vs. 81.6%; 2-year DFS, 80.7 vs. 42.9%; P=0.047), while no significant differences were observed in overall survival (OS; 1-year OS, 100.0 vs. 95.7%; 2-year OS, 90.0 vs. 64.9%; P=0.187). Additionally, the majority of adverse event incidences (apart from leukopenia) did not differ significantly between the ICI + chemo and chemo groups (all P>0.050). On the whole, the present study demonstrated that neoadjuvant ICI treatment + chemotherapy exhibited adequate efficacy and acceptable toxicity compared with chemotherapy alone in patients with locally advanced NSCLC.
Collapse
Affiliation(s)
- Yi Yang
- Department of Thoracic Surgery, The Third People's Hospital of Chengdu, Chengdu, Sichuan 610082, P.R. China
| | - Zaoyang Liu
- Department of Thoracic Surgery, The Third People's Hospital of Chengdu, Chengdu, Sichuan 610082, P.R. China
| |
Collapse
|
13
|
Wang Y, Huang S, Feng X, Xu W, Luo R, Zhu Z, Zeng Q, He Z. Advances in efficacy prediction and monitoring of neoadjuvant immunotherapy for non-small cell lung cancer. Front Oncol 2023; 13:1145128. [PMID: 37265800 PMCID: PMC10229830 DOI: 10.3389/fonc.2023.1145128] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 05/03/2023] [Indexed: 06/03/2023] Open
Abstract
The use of immune checkpoint inhibitors (ICIs) has become mainstream in the treatment of non-small cell lung cancer (NSCLC). The idea of harnessing the immune system to fight cancer is fast developing. Neoadjuvant treatment in NSCLC is undergoing unprecedented change. Chemo-immunotherapy combinations not only seem to achieve population-wide treating coverage irrespective of PD-L1 expression but also enable achieving a pathological complete response (pCR). Despite these recent advancements in neoadjuvant chemo-immunotherapy, not all patients respond favorably to treatment with ICIs plus chemo and may even suffer from severe immune-related adverse effects (irAEs). Similar to selection for target therapy, identifying patients most likely to benefit from chemo-immunotherapy may be valuable. Recently, several prognostic and predictive factors associated with the efficacy of neoadjuvant immunotherapy in NSCLC, such as tumor-intrinsic biomarkers, tumor microenvironment biomarkers, liquid biopsies, microbiota, metabolic profiles, and clinical characteristics, have been described. However, a specific and sensitive biomarker remains to be identified. Recently, the construction of prediction models for ICI therapy using novel tools, such as multi-omics factors, proteomic tests, host immune classifiers, and machine learning algorithms, has gained attention. In this review, we provide a comprehensive overview of the different positive prognostic and predictive factors in treating preoperative patients with ICIs, highlight the recent advances made in the efficacy prediction of neoadjuvant immunotherapy, and provide an outlook for joint predictors.
Collapse
Affiliation(s)
- Yunzhen Wang
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Sha Huang
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiangwei Feng
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wangjue Xu
- Department of Thoracic Surgery, Longyou County People’s Hospital, Longyou, China
| | - Raojun Luo
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ziyi Zhu
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qingxin Zeng
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhengfu He
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
14
|
Cabezas-Camarero S, García-Barberán V, Benítez-Fuentes JD, Sotelo MJ, Plaza JC, Encinas-Bascones A, De-la-Sen Ó, Falahat F, Gimeno-Hernández J, Gómez-Serrano M, Puebla-Díaz F, De-Pedro-Marina M, Iglesias-Moreno M, Pérez-Segura P. Clinical Behavior, Mutational Profile and T-Cell Repertoire of High-Grade Neuroendocrine Tumors of the Head and Neck. Cancers (Basel) 2023; 15:cancers15092431. [PMID: 37173898 PMCID: PMC10177201 DOI: 10.3390/cancers15092431] [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: 03/15/2023] [Revised: 04/15/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Neuroendocrine carcinomas (NECs) of the head and neck (HN) account for <1% of HN cancers (HNCs), with a 5-year overall survival (OS) <20%. This is a retrospective study of HN NECs diagnosed at our institution between 2005 and 2022. Immunohistochemistry and next-generation sequencing (NGS) were used to evaluate neuroendocrine markers, tumor mutational burden (TMB), mutational profiles and T-cell receptor repertoires. Eleven patients with high-grade HN NECs were identified (male:female ratio 6:5; median age 61 (Min-Max: 31-86)): nasoethmoidal (3), parotid gland (3), submaxillary gland (1), larynx (3) and base of tongue (1). Among n = 8 stage II/IVA/B, all received (chemo)radiotherapy with/without prior surgery or induction chemotherapy, with complete response in 7/8 (87.5%). Among n = 6 recurrent/metastatic patients, three received anti-PD1 (nivolumab (2), pembrolizumab (1)): two achieved partial responses lasting 24 and 10 months. After a median follow-up of 30 and 23.5 months since diagnosis and since recurrent/metastatic, median OS was not reached. Median TMB (n = 7) was 6.72 Mut/Mb. The most common pathogenic variants were TP53, HNF1A, SMARCB1, CDKN2A, PIK3CA, RB1 and MYC. There were 224 median TCR clones (n = 5 pts). In one patient, TCR clones increased from 59 to 1446 after nivolumab. HN NECs may achieve long-lasting survival with multimodality treatment. They harbor moderate-high TMBs and large TCR repertoires, which may explain responses to anti-PD1 agents in two patients and justify the study of immunotherapy in this disease.
Collapse
Affiliation(s)
- Santiago Cabezas-Camarero
- Medical Oncology Department, Hospital Clínico Universitario San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain
| | - Vanesa García-Barberán
- Molecular Oncology Laboratory, Hospital Clínico Universitario San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain
| | - Javier David Benítez-Fuentes
- Medical Oncology Department, Hospital Clínico Universitario San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain
| | - Miguel J Sotelo
- Medical Oncology Department, Aliada Cancer Center, Lima 15036, Peru
- Medical Oncology Department, Clínica San Felipe, Lima 15072, Peru
- Medical Oncology Department, Hospital María Auxiliadora, Lima 15801, Peru
| | - José Carlos Plaza
- Pathology Department, Hospital Clínico Universitario San Carlos, 28040 Madrid, Spain
| | | | - Óscar De-la-Sen
- Maxillofacial Surgery Department, Hospital Clínico Universitario San Carlos, 28040 Madrid, Spain
| | - Farzin Falahat
- Maxillofacial Surgery Department, Hospital Clínico Universitario San Carlos, 28040 Madrid, Spain
| | - Jesús Gimeno-Hernández
- Otolaryngology-Head and Neck Surgery Department, Hospital Clínico Universitario San Carlos, 28040 Madrid, Spain
| | - Manuel Gómez-Serrano
- Otolaryngology-Head and Neck Surgery Department, Hospital Clínico Universitario San Carlos, 28040 Madrid, Spain
| | - Fernando Puebla-Díaz
- Radiation Oncology Department, Hospital Clínico Universitario San Carlos, 28040 Madrid, Spain
| | - Manuel De-Pedro-Marina
- Maxillofacial Surgery Department, Hospital Clínico Universitario San Carlos, 28040 Madrid, Spain
| | - Maricruz Iglesias-Moreno
- Maxillofacial Surgery Department, Hospital Clínico Universitario San Carlos, 28040 Madrid, Spain
| | - Pedro Pérez-Segura
- Medical Oncology Department, Hospital Clínico Universitario San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain
| |
Collapse
|
15
|
Qi T, Vincent BG, Cao Y. A multispecies framework for modeling adaptive immunity and immunotherapy in cancer. PLoS Comput Biol 2023; 19:e1010976. [PMID: 37083574 PMCID: PMC10155959 DOI: 10.1371/journal.pcbi.1010976] [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: 07/06/2022] [Revised: 05/03/2023] [Accepted: 02/24/2023] [Indexed: 04/22/2023] Open
Abstract
Predator-prey theory is commonly used to describe tumor growth in the presence of selective pressure from the adaptive immune system. These interactions are mediated by the tumor immunopeptidome (what the tumor "shows" the body) and the T-cell receptor (TCR) repertoire (how well the body "sees" cancer cells). The tumor immunopeptidome comprises neoantigens which can be gained and lost throughout tumorigenesis and treatment. Heterogeneity in the immunopeptidome is predictive of poor response to immunotherapy in some tumor types, suggesting that the TCR repertoire is unable to support a fully polyclonal response against every neoantigen. Importantly, while tumor and T-cell populations are known to compete with each other for intratumoral resources, whether between-lineage competition among peripheral T cells influences the TCR repertoire is unknown and difficult to interrogate experimentally. Computational models may offer a way to investigate these phenomena and deepen our understanding of the tumor-immune axis. Here, we construct a predator-prey-like model and calibrate it to preclinical and clinical data to describe tumor growth and immunopeptidome diversification. Simultaneously, we model the expansion of antigen-specific T-cell lineages and their consumption of both lineage-specific antigenic resources and lineage-agnostic, shared resources. This predator-prey-like framework accurately described clinically observed immunopeptidomes; recapitulated response-associated effects of immunotherapy, including immunoediting; and allowed exploration of treatment of tumors with varying growth and mutation rates.
Collapse
Affiliation(s)
- Timothy Qi
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Benjamin G. Vincent
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Division of Hematology/Oncology, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Yanguang Cao
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| |
Collapse
|
16
|
Li C, Wu J, Jiang L, Zhang L, Huang J, Tian Y, Zhao Y, Liu X, Xia L, E H, Gao P, Hou L, Yang M, Ma M, Su C, Zhang H, Chen H, She Y, Xie D, Luo Q, Chen C. The predictive value of inflammatory biomarkers for major pathological response in non-small cell lung cancer patients receiving neoadjuvant chemoimmunotherapy and its association with the immune-related tumor microenvironment: a multi-center study. Cancer Immunol Immunother 2023; 72:783-794. [PMID: 36056951 DOI: 10.1007/s00262-022-03262-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 07/19/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND Inflammatory biomarkers in the peripheral blood have been established as predictors for immunotherapeutic efficacy in advanced non-small cell lung cancer (NSCLC). Whether they can also predict major pathological response (MPR) in neoadjuvant setting remains unclear. METHODS In this multi-center retrospective study, 122 and 92 stage I-IIIB NSCLC patients from six hospitals who received neoadjuvant chemoimmunotherapy followed by surgery were included in the discovery and external validation cohort, respectively. Baseline and on-treatment neutrophil-to-lymphocyte ratio (NLR), derived NLR (dNLR), platelet-to-lymphocyte ratio (PLR), monocyte-to-lymphocyte ratio (MLR) and systemic immune-inflammation index (SII) were calculated and associated with MPR. Furthermore, resected tumor samples from 37 patients were collected for RNA-sequencing to investigate the immune-related tumor microenvironment. RESULTS In both the discovery and validation cohorts, the on-treatment NLR, dNLR, PLR, and SII levels were significantly lower in the patients with MPR versus non-MPR. On-treatment SII remained an independent predictor of MPR in multivariate logistic regression analysis. The area under the curve (AUC) of on-treatment SII for predicting MPR was 0.75 (95%CI, 0.67-0.84) in the discovery cohort. Moreover, the predictive value was further improved by combining the on-treatment SII and radiological tumor regression data, demonstrating an AUC of 0.82 (95%CI, 0.74-0.90). The predictive accuracy was validated in the external cohort. Compared with the SII-high group, patients with SII-Low were associated with the activated B cell receptor signaling pathway and a higher intratumoral immune cell infiltration level. CONCLUSIONS On-treatment SII was independently associated with MPR in NSCLC patients receiving neoadjuvant chemoimmunotherapy. Further prospective studies are warranted.
Collapse
Affiliation(s)
- Chongwu Li
- Department of Thoracic Surgery, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, People's Republic of China
| | - Junqi Wu
- Department of Thoracic Surgery, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, People's Republic of China
| | - Long Jiang
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Lei Zhang
- Department of Thoracic Surgery, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, People's Republic of China
| | - Jia Huang
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Yu Tian
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Yue Zhao
- Department of Thoracic Surgery, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, People's Republic of China
| | - Xiucheng Liu
- Department of Thoracic Surgery, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, People's Republic of China
| | - Lang Xia
- Department of Thoracic Surgery, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, People's Republic of China
| | - Haoran E
- Department of Thoracic Surgery, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, People's Republic of China
| | - Peigen Gao
- Department of Thoracic Surgery, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, People's Republic of China
| | - Likun Hou
- Department of Pathology, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, People's Republic of China
| | - Minglei Yang
- Department of Thoracic Surgery, Ningbo No. 2 Hospital, Chinese Academy of Sciences, Zhejiang, People's Republic of China
| | - Minjie Ma
- Department of Thoracic Surgery, The First Hospital of Lanzhou University, Gansu, People's Republic of China
| | - Chunxia Su
- Department of Oncology, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, People's Republic of China
| | - Hao Zhang
- Department of Thoracic Surgery, Affiliated Hospital of Xuzhou Medical University, Jiangsu, People's Republic of China
| | - Hezhong Chen
- Department of Thoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, People's Republic of China
| | - Yunlang She
- Department of Thoracic Surgery, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, People's Republic of China
| | - Dong Xie
- Department of Thoracic Surgery, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, People's Republic of China.
| | - Qingquan Luo
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, People's Republic of China.
| | - Chang Chen
- Department of Thoracic Surgery, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, People's Republic of China.
| |
Collapse
|
17
|
Shin J, Ham B, Seo JH, Lee SB, Park IA, Gong G, Kim SB, Lee HJ. Immune repertoire and responses to neoadjuvant TCHP therapy in HER2-positive breast cancer. Ther Adv Med Oncol 2023; 15:17588359231157654. [PMID: 36865681 PMCID: PMC9972050 DOI: 10.1177/17588359231157654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 01/30/2023] [Indexed: 03/02/2023] Open
Abstract
Background Despite the introduction of trastuzumab, pathologic complete response (pCR) is not attained in approximately 30-40% of Human epithelial growth factor receptor-2-positive breast cancer. Tumor-infiltrating lymphocytes (TIL) have been suggested as a predictive marker of treatment response, albeit not always effective. We investigated the relationship between trastuzumab, docetaxel, carboplatin, and pertuzumab (TCHP) treatment and immune repertoire as a treatment response predictor. Design In all, 35 cases were divided into two experimental groups: 10 and 25 cases in the preliminary and main experiments, respectively. In the preliminary experiment, the biopsy tissues before TCHP treatment and the surgical tissues after TCHP treatment were compared. In the main experiment, the biopsy tissues before TCHP treatment were compared according to the TCHP treatment response. Methods The T-cell repertoire for TRA, TRB, TRG, and TRD, and B-cell repertoire for immunoglobulin heavy, immunoglobulin kappa, and immunoglobulin lambda were evaluated. Whole transcriptome sequencing was also performed. Results In the preliminary experiment, the density and richness of the T-cell receptor (TCR) and B-cell receptor (BCR) repertoires decreased after treatment, regardless of TCHP response. In the main experiment, the Shannon's entropy index, density, and length of CDR3 of the TCR and BCR repertoires did not differ significantly in patients who did and did not achieve pCR. The pCR and non-pCR subgroups according to the level of TILs revealed that the non-pCR/lowTIL group had a higher proportion of low-frequency clones than the pCR/lowTIL group in TRA (non-pCR/lowTIL versus pCR/lowTIL, 0.01-0.1%, 63% versus 45.3%; <0.01%, 32.9% versus 51.8%, p < 0.001) and TRB (non-pCR/lowTIL versus pCR/lowTIL, 0.01-0.1%, 26.5% versus 14.7%; <0.01%, 72.0% versus 84.1%, p < 0.001). Conclusions The role of the diversity, richness, and density of the TCR and BCR repertoires as predictive markers for TCHP response was not identified. Compositions of low-frequency clones could be candidates for predictive factors of TCHP response; however, validation studies and further research are necessary.
Collapse
Affiliation(s)
- Junyoung Shin
- Department of Pathology, Asan Medical Center,
University of Ulsan College of Medicine, Seoul, Korea
| | | | | | - Sae Byul Lee
- Department of Breast Surgery, Asan Medical
Center, University of Ulsan College of Medicine, Seoul, Korea
| | - In Ah Park
- Department of Pathology, Kangbuk Samsung
Hospital, Seoul, Republic of Korea
| | - Gyungyub Gong
- Department of Pathology, Asan Medical Center,
University of Ulsan College of Medicine, Seoul, Korea
| | | | | |
Collapse
|
18
|
Peng L, Bin Y, Ding P, Chen L, Zeng H, Xu Z, Ji L, Gao X, Liu P, Wang Y, Zhang S, Liao Z, Xia X, Zhang R, Tong F, Dong X. Integrated circulating tumor DNA and T cell repertoire predict radiotherapeutic response and outcome in non-small cell lung cancer patients with brain metastasis. Cancer Commun (Lond) 2023; 43:625-629. [PMID: 36815673 PMCID: PMC10174081 DOI: 10.1002/cac2.12410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 11/07/2022] [Accepted: 02/02/2023] [Indexed: 02/24/2023] Open
Affiliation(s)
- Ling Peng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Yawen Bin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Peng Ding
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Lingjuan Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Hao Zeng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Zelong Xu
- Geneplus-Beijing, Beijing, P. R. China
| | - Liyan Ji
- Geneplus-Beijing, Beijing, P. R. China
| | - Xuan Gao
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P. R. China.,GenePlus-Shenzhen Clinical Laboratory, Shenzhen, Guangdong, P. R. China
| | - Pian Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Ye Wang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Sheng Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Zhongxing Liao
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Ruiguang Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Fan Tong
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Xiaorong Dong
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| |
Collapse
|
19
|
Roulleaux Dugage M, Albarrán-Artahona V, Laguna JC, Chaput N, Vignot S, Besse B, Mezquita L, Auclin E. Biomarkers of response to immunotherapy in early stage non-small cell lung cancer. Eur J Cancer 2023; 184:179-196. [PMID: 36963241 DOI: 10.1016/j.ejca.2023.01.029] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 01/30/2023] [Indexed: 02/19/2023]
Abstract
Immunotherapy with immune-checkpoint inhibitors (ICIs) targeting programmed cell death 1 or programmed death-ligand 1 has revolutionised the treatment of advanced non-small cell lung cancer (NSCLC) and has been investigated in early NSCLC, alone or in combination with chemotherapy, anti-CTLA-4 antibodies and radiotherapy. Although more mature data are needed before setting a change of paradigm in early stages, reports of pathological response rates and disease-free survival are promising, especially with neoadjuvant multimodality approaches. Nevertheless, major pathological response rates for neoadjuvant anti-PD-(L)1 monotherapy rarely exceed 40%, and biomarkers for characterising patients who may benefit the most from ICIs are lacking. These biomarkers have a distinct value from the metastatic setting, with highly different tumour biologies. Among the most investigated so far in this context, programmed death-ligand 1 expression and, to a lesser extent, tumour mutational burden seem to correlate better with higher pathological response rates and survival. Epidermal growth factor receptor, Serine/Threonine Kinase 11and Kelch-like ECH-associated protein 1 mutations rise as essential determinations for the treatment selection in early-stage NSCLC. Emerging and promising approaches comprise evaluation of blood-based ratios, microbiota, and baseline intratumoural TCR clonality. Circulating tumour DNA will be of great help in the near future when selecting best candidates for adjuvant ICIs, monitoring the tumour response to the neoadjuvant treatment in order to improve the rates of complete resections in the early stage.
Collapse
Affiliation(s)
- Matthieu Roulleaux Dugage
- Department of Oncology, Hôpital Européen Georges Pompidou, AP-HP, Université Paris Cité, Paris, France; Laboratoire D'Immunomonitoring en Oncologie, INSERM US23, CNRS UMS 3655, Gustave Roussy, Villejuif, Île-de-France, France
| | - Víctor Albarrán-Artahona
- Medical Oncology Department, Hospital Clinic de Barcelona, Spain; Laboratory of Translational Genomics and Targeted Therapies in Solid Tumors, IDIBAPS, Barcelona, Spain
| | | | - Nathalie Chaput
- Laboratoire D'Immunomonitoring en Oncologie, INSERM US23, CNRS UMS 3655, Gustave Roussy, Villejuif, Île-de-France, France
| | | | - Benjamin Besse
- Department of Oncology, Gustave Roussy, Villejuif, Île-de-France, France
| | - Laura Mezquita
- Medical Oncology Department, Hospital Clinic de Barcelona, Spain; Laboratory of Translational Genomics and Targeted Therapies in Solid Tumors, IDIBAPS, Barcelona, Spain; Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Edouard Auclin
- Department of Oncology, Hôpital Européen Georges Pompidou, AP-HP, Université Paris Cité, Paris, France.
| |
Collapse
|
20
|
Shang J, Zhu X, Sun Y, Li F, Kong X, Liu JX. DM-MOGA: a multi-objective optimization genetic algorithm for identifying disease modules of non-small cell lung cancer. BMC Bioinformatics 2023; 24:13. [PMID: 36624376 PMCID: PMC9830734 DOI: 10.1186/s12859-023-05136-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Constructing molecular interaction networks from microarray data and then identifying disease module biomarkers can provide insight into the underlying pathogenic mechanisms of non-small cell lung cancer. A promising approach for identifying disease modules in the network is community detection. RESULTS In order to identify disease modules from gene co-expression networks, a community detection method is proposed based on multi-objective optimization genetic algorithm with decomposition. The method is named DM-MOGA and possesses two highlights. First, the boundary correction strategy is designed for the modules obtained in the process of local module detection and pre-simplification. Second, during the evolution, we introduce Davies-Bouldin index and clustering coefficient as fitness functions which are improved and migrated to weighted networks. In order to identify modules that are more relevant to diseases, the above strategies are designed to consider the network topology of genes and the strength of connections with other genes at the same time. Experimental results of different gene expression datasets of non-small cell lung cancer demonstrate that the core modules obtained by DM-MOGA are more effective than those obtained by several other advanced module identification methods. CONCLUSIONS The proposed method identifies disease-relevant modules by optimizing two novel fitness functions to simultaneously consider the local topology of each gene and its connection strength with other genes. The association of the identified core modules with lung cancer has been confirmed by pathway and gene ontology enrichment analysis.
Collapse
Affiliation(s)
- Junliang Shang
- grid.412638.a0000 0001 0227 8151School of Computer Science, Qufu Normal University, Rizhao, 276826 China
| | - Xuhui Zhu
- grid.412638.a0000 0001 0227 8151School of Computer Science, Qufu Normal University, Rizhao, 276826 China
| | - Yan Sun
- grid.412638.a0000 0001 0227 8151School of Computer Science, Qufu Normal University, Rizhao, 276826 China
| | - Feng Li
- grid.412638.a0000 0001 0227 8151School of Computer Science, Qufu Normal University, Rizhao, 276826 China
| | - Xiangzhen Kong
- grid.412638.a0000 0001 0227 8151School of Computer Science, Qufu Normal University, Rizhao, 276826 China
| | - Jin-Xing Liu
- grid.412638.a0000 0001 0227 8151School of Computer Science, Qufu Normal University, Rizhao, 276826 China
| |
Collapse
|
21
|
Liu C, Zhao W, Xie J, Lin H, Hu X, Li C, Shang Y, Wang Y, Jiang Y, Ding M, Peng M, Xu T, Hu A, Huang Y, Gao Y, Liu X, Liu J, Ma F. Development and validation of a radiomics-based nomogram for predicting a major pathological response to neoadjuvant immunochemotherapy for patients with potentially resectable non-small cell lung cancer. Front Immunol 2023; 14:1115291. [PMID: 36875128 PMCID: PMC9978193 DOI: 10.3389/fimmu.2023.1115291] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 02/02/2023] [Indexed: 02/18/2023] Open
Abstract
Introduction The treatment response to neoadjuvant immunochemotherapy varies among patients with potentially resectable non-small cell lung cancers (NSCLC) and may have severe immune-related adverse effects. We are currently unable to accurately predict therapeutic response. We aimed to develop a radiomics-based nomogram to predict a major pathological response (MPR) of potentially resectable NSCLC to neoadjuvant immunochemotherapy using pretreatment computed tomography (CT) images and clinical characteristics. Methods A total of 89 eligible participants were included and randomly divided into training (N=64) and validation (N=25) sets. Radiomic features were extracted from tumor volumes of interest in pretreatment CT images. Following data dimension reduction, feature selection, and radiomic signature building, a radiomics-clinical combined nomogram was developed using logistic regression analysis. Results The radiomics-clinical combined model achieved excellent discriminative performance, with AUCs of 0.84 (95% CI, 0.74-0.93) and 0.81(95% CI, 0.63-0.98) and accuracies of 80% and 80% in the training and validation sets, respectively. Decision curves analysis (DCA) indicated that the radiomics-clinical combined nomogram was clinically valuable. Discussion The constructed nomogram was able to predict MPR to neoadjuvant immunochemotherapy with a high degree of accuracy and robustness, suggesting that it is a convenient tool for assisting with the individualized management of patients with potentially resectable NSCLC.
Collapse
Affiliation(s)
- Chaoyuan Liu
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei Zhao
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Clinical Research Center for Medical Imaging in Hunan Province, Changsha, Hunan, China
| | - Junpeng Xie
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Huashan Lin
- Department of Pharmaceutical Diagnosis, GE Healthcare, Changsha, China
| | - Xingsheng Hu
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chang Li
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Youlan Shang
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yapeng Wang
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yingjia Jiang
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Mengge Ding
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Muyun Peng
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Tian Xu
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ao'ran Hu
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuda Huang
- Department of Ministry of science and technology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuan Gao
- Department of Basic Science, College of Chiropractic, Logan University, Chester field, MO, United States
| | - Xianling Liu
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jun Liu
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Clinical Research Center for Medical Imaging in Hunan Province, Changsha, Hunan, China.,Radiology Quality Control Center, Changsha, Hunan, China
| | - Fang Ma
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
22
|
Høye E, Dagenborg VJ, Torgunrud A, Lund-Andersen C, Fretland ÅA, Lorenz S, Edwin B, Hovig E, Fromm B, Inderberg EM, Greiff V, Ree AH, Flatmark K. T cell receptor repertoire sequencing reveals chemotherapy-driven clonal expansion in colorectal liver metastases. Gigascience 2022; 12:giad032. [PMID: 37161965 PMCID: PMC10170408 DOI: 10.1093/gigascience/giad032] [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: 09/30/2022] [Revised: 02/07/2023] [Accepted: 04/25/2023] [Indexed: 05/11/2023] Open
Abstract
BACKGROUND Colorectal liver metastasis (CLM) is a leading cause of colorectal cancer mortality, and the response to immune checkpoint inhibition (ICI) in microsatellite-stable CRC has been disappointing. Administration of cytotoxic chemotherapy may cause increased density of tumor-infiltrating T cells, which has been associated with improved response to ICI. This study aimed to quantify and characterize T-cell infiltration in CLM using T-cell receptor (TCR) repertoire sequencing. Eighty-five resected CLMs from patients included in the Oslo CoMet study were subjected to TCR repertoire sequencing. Thirty-five and 15 patients had received neoadjuvant chemotherapy (NACT) within a short or long interval, respectively, prior to resection, while 35 patients had not been exposed to NACT. T-cell fractions were calculated, repertoire clonality was analyzed based on Hill evenness curves, and TCR sequence convergence was assessed using network analysis. RESULTS Increased T-cell fractions (10.6% vs. 6.3%) were detected in CLMs exposed to NACT within a short interval prior to resection, while modestly increased clonality was observed in NACT-exposed tumors independently of the timing of NACT administration and surgery. While private clones made up >90% of detected clones, network connectivity analysis revealed that public clones contributed the majority of TCR sequence convergence. CONCLUSIONS TCR repertoire sequencing can be used to quantify T-cell infiltration and clonality in clinical samples. This study provides evidence to support chemotherapy-driven T-cell clonal expansion in CLM in a clinical context.
Collapse
Affiliation(s)
- Eirik Høye
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
- Institute of Clinical Medicine, Medical Faculty, University of Oslo, 0318 Oslo, Norway
| | - Vegar J Dagenborg
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
- Department of Gastroenterological Surgery, The Norwegian Radium Hospital, 0379 Oslo, Norway
| | - Annette Torgunrud
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
| | - Christin Lund-Andersen
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
- Institute of Clinical Medicine, Medical Faculty, University of Oslo, 0318 Oslo, Norway
| | - Åsmund A Fretland
- The Intervention Centre, Rikshospitalet, Oslo University Hospital, 0372 Oslo, Norway
- Department of Hepato-Pancreato-Biliary Surgery, Rikshospitalet, Oslo University Hospital, 0372 Oslo, Norway
| | - Susanne Lorenz
- Department of Core Facilities, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
| | - Bjørn Edwin
- Institute of Clinical Medicine, Medical Faculty, University of Oslo, 0318 Oslo, Norway
- The Intervention Centre, Rikshospitalet, Oslo University Hospital, 0372 Oslo, Norway
- Department of Hepato-Pancreato-Biliary Surgery, Rikshospitalet, Oslo University Hospital, 0372 Oslo, Norway
| | - Eivind Hovig
- Center for Bioinformatics, Department of Informatics, University of Oslo, 0316 Oslo, Norway
| | - Bastian Fromm
- The Arctic University Museum of Norway, UiT – The Arctic University of Norway, 9037 Tromsø, Norway
| | - Else M Inderberg
- Translational Research Unit, Department of Cellular Therapy, Oslo University Hospital, 0379 Oslo, Norway
| | - Victor Greiff
- Department of Immunology, University of Oslo and Oslo University Hospital, 0372 Oslo, Norway
| | - Anne H Ree
- Institute of Clinical Medicine, Medical Faculty, University of Oslo, 0318 Oslo, Norway
- Department of Oncology, Akershus University Hospital, 1478 Lørenskog, Norway
| | - Kjersti Flatmark
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
- Institute of Clinical Medicine, Medical Faculty, University of Oslo, 0318 Oslo, Norway
- Department of Gastroenterological Surgery, The Norwegian Radium Hospital, 0379 Oslo, Norway
| |
Collapse
|
23
|
Porciello N, Franzese O, D’Ambrosio L, Palermo B, Nisticò P. T-cell repertoire diversity: friend or foe for protective antitumor response? JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:356. [PMID: 36550555 PMCID: PMC9773533 DOI: 10.1186/s13046-022-02566-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022]
Abstract
Profiling the T-Cell Receptor (TCR) repertoire is establishing as a potent approach to investigate autologous and treatment-induced antitumor immune response. Technical and computational breakthroughs, including high throughput next-generation sequencing (NGS) approaches and spatial transcriptomics, are providing unprecedented insight into the mechanisms underlying antitumor immunity. A precise spatiotemporal variation of T-cell repertoire, which dynamically mirrors the functional state of the evolving host-cancer interaction, allows the tracking of the T-cell populations at play, and may identify the key cells responsible for tumor eradication, the evaluation of minimal residual disease and the identification of biomarkers of response to immunotherapy. In this review we will discuss the relationship between global metrics characterizing the TCR repertoire such as T-cell clonality and diversity and the resultant functional responses. In particular, we will explore how specific TCR repertoires in cancer patients can be predictive of prognosis or response to therapy and in particular how a given TCR re-arrangement, following immunotherapy, can predict a specific clinical outcome. Finally, we will examine current improvements in terms of T-cell sequencing, discussing advantages and challenges of current methodologies.
Collapse
Affiliation(s)
- Nicla Porciello
- grid.417520.50000 0004 1760 5276Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Ornella Franzese
- grid.6530.00000 0001 2300 0941Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Lorenzo D’Ambrosio
- grid.417520.50000 0004 1760 5276Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Belinda Palermo
- grid.417520.50000 0004 1760 5276Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Paola Nisticò
- grid.417520.50000 0004 1760 5276Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
24
|
Wang Y, Peng L, Zhao M, Xiong Y, Xue J, Li B, Huang Z, Liu X, Yang X, Song Y, Bing Z, Guo C, Tian Z, Gao C, Cao L, Cao Z, Li J, Jiang X, Si X, Zhang L, Li X, Zheng Z, Song M, Chen R, Lim WT, Pavan A, Romero A, Liang N, Yang H, Li S. Comprehensive analysis of T cell receptor repertoire in patients with KRAS mutant non-small cell lung cancer. Transl Lung Cancer Res 2022; 11:1936-1950. [PMID: 36248331 PMCID: PMC9554687 DOI: 10.21037/tlcr-22-629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 09/14/2022] [Indexed: 11/06/2022]
Abstract
Background Kirsten rat sarcoma viral oncogene homolog (KRAS) is one of the most frequently mutated oncogenes in non-small cell lung cancer (NSCLC). The administration of immunotherapy has demonstrated significant efficacy in prolonging the overall survival of patients with KRAS mutation in recent years. However, the efficacy of immunotherapy in KRAS mutant NSCLC is variable. Analysis of T cell receptor (TCR) repertoire may contribute to a better understanding of the mechanisms behind such differential outcomes. Methods A total of 47 patients with KRAS mutant NSCLC were enrolled in this study. Deep sequencing of the TCR β chain complementarity-determining regions in tumor tissue and paired peripheral blood specimens was conducted. Comprehensive analysis of TCR repertoire metrics was performed with different KRAS mutation subtypes and concomitant mutations. Moreover, the associations between TCR repertoire metrics and tumor mutation burden (TMB), as well as programmed death-ligand 1 were explored, respectively. Results TCR repertoire metrics, including Shannon index, Clonality, and Morisita index (MOI), showed no significant differences among different KRAS mutation subtypes. The similar results were observed between patients with tumor protein p53 (TP53) mutation and those with wild-type TP53. In contrast, although no significant differences were found in Shannon index and Clonality, patients with KRAS/serine/threonine kinase 11 (STK11) comutation showed a significantly higher MOI compared to their STK11 wild-type counterparts (P=0.012). In addition, TCR repertoire metrics were neither associated with TMB nor programmed death-ligand 1 expression in KRAS mutant NSCLC. Conclusions This retrospective study comprehensively described the TCR repertoire in KRAS mutant NSCLC. A higher MOI represented more overlap of the TCR repertoire between tumor tissue and paired peripheral blood, indicating distinctive immunological features in NSCLC with KRAS/STK11 comutation.
Collapse
Affiliation(s)
- Yadong Wang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ling Peng
- Department of Respiratory Disease, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Ming Zhao
- Department of Thoracic Surgery, the General Hospital of the People’s Liberation Army, Beijing, China
| | | | - Jianchao Xue
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bowen Li
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhicheng Huang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xinyu Liu
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Eight-Year MD Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaoying Yang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Eight-Year MD Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yang Song
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhongxing Bing
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chao Guo
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhenhuan Tian
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chao Gao
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lei Cao
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhili Cao
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ji Li
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xu Jiang
- Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaoyan Si
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li Zhang
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaoguang Li
- Minimally Invasive Tumor Therapies Center, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhibo Zheng
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of International Medical Services, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | | | | | - Wan-Teck Lim
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Alberto Pavan
- Medical Oncology Department, AULSS 3 Serenissima, Mestre-Venezia, Italy
| | - Atocha Romero
- Medical Oncology Department, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Naixin Liang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Huaxia Yang
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Rheumatology and Clinical Immunology, National Clinical Research Center for Dermatologic and Immunologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, the Ministry of Education Key Laboratory, Beijing, China
| | - Shanqing Li
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
25
|
Casarrubios M, Provencio M, Nadal E, Insa A, Del Rosario García-Campelo M, Lázaro-Quintela M, Dómine M, Majem M, Rodriguez-Abreu D, Martinez-Marti A, De Castro Carpeño J, Cobo M, López Vivanco G, Del Barco E, Bernabé R, Viñolas N, Barneto Aranda I, Massuti B, Sierra-Rodero B, Martinez-Toledo C, Fernández-Miranda I, Serna-Blanco R, Romero A, Calvo V, Cruz-Bermúdez A. Tumor microenvironment gene expression profiles associated to complete pathological response and disease progression in resectable NSCLC patients treated with neoadjuvant chemoimmunotherapy. J Immunother Cancer 2022; 10:jitc-2022-005320. [PMID: 36171009 PMCID: PMC9528578 DOI: 10.1136/jitc-2022-005320] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2022] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Neoadjuvant chemoimmunotherapy for non-small cell lung cancer (NSCLC) has improved pathological responses and survival rates compared with chemotherapy alone, leading to Food and Drug Administration (FDA) approval of nivolumab plus chemotherapy for resectable stage IB-IIIA NSCLC (AJCC 7th edition) without ALK or EGFR alterations. Unfortunately, a considerable percentage of tumors do not completely respond to therapy, which has been associated with early disease progression. So far, it is impossible to predict these events due to lack of knowledge. In this study, we characterized the gene expression profile of tumor samples to identify new biomarkers and mechanisms behind tumor responses to neoadjuvant chemoimmunotherapy and disease recurrence after surgery. METHODS Tumor bulk RNA sequencing was performed in 16 pretreatment and 36 post-treatment tissue samples from 41 patients with resectable stage IIIA NSCLC treated with neoadjuvant chemoimmunotherapy from NADIM trial. A panel targeting 395 genes related to immunological processes was used. Tumors were classified as complete pathological response (CPR) and non-CPR, based on the total absence of viable tumor cells in tumor bed and lymph nodes tested at surgery. Differential-expressed genes between groups and pathway enrichment analysis were assessed using DESeq2 and gene set enrichment analysis. CIBERSORTx was used to estimate the proportions of immune cell subtypes. RESULTS CPR tumors had a stronger pre-established immune infiltrate at baseline than non-CPR, characterized by higher levels of IFNG, GZMB, NKG7, and M1 macrophages, all with a significant area under the receiver operating characteristic curve (ROC) >0.9 for CPR prediction. A greater effect of neoadjuvant therapy was also seen in CPR tumors with a reduction of tumor markers and IFNγ signaling after treatment. Additionally, the higher expression of several genes, including AKT1, BST2, OAS3, or CD8B; or higher dendritic cells and neutrophils proportions in post-treatment non-CPR samples, were associated with relapse after surgery. Also, high pretreatment PD-L1 and tumor mutational burden levels influenced the post-treatment immune landscape with the downregulation of proliferation markers and type I interferon signaling molecules in surgery samples. CONCLUSIONS Our results reinforce the differences between CPR and non-CPR responses, describing possible response and relapse immune mechanisms, opening the possibility of therapy personalization of immunotherapy-based regimens in the neoadjuvant setting of NSCLC.
Collapse
Affiliation(s)
- Marta Casarrubios
- Medical Oncology, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDIPHISA), Hospital Universitario Puerta de Hierro-Majadahonda, Majadahonda-Madrid, Spain
| | - Mariano Provencio
- Medical Oncology, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDIPHISA), Hospital Universitario Puerta de Hierro-Majadahonda, Majadahonda-Madrid, Spain
| | - Ernest Nadal
- Medical Oncology, Catalan Institute of Oncology, Oncobell Program, IDIBELL, L'Hospitalet de Llobregat, L'Hospitalet, Barcelona, Spain
| | - Amelia Insa
- Medical Oncology, Fundación INCLIVA, Hospital Clínico Universitario de Valencia, Valencia, Spain
| | | | | | - Manuel Dómine
- Medical Oncology, Hospital Universitario Fundación Jiménez Díaz, Madrid, Spain
| | - Margarita Majem
- Medical Oncology, Hospital de la Santa Creu i Sant Pau Servei de Oncologia Medica, Barcelona, Spain
| | - Delvys Rodriguez-Abreu
- Medical Oncology, Hospital Universitario Insular de Gran Canaria, Las Palmas, Canarias, Spain
| | - Alex Martinez-Marti
- Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, Barcelona, Spain, Barcelona, Barcelona, Spain
| | | | - Manuel Cobo
- Medical Oncology Intercenter Unit, Regional and Virgen de la Victoria University Hospitals, IBIMA, Málaga, Spain
| | | | - Edel Del Barco
- Medical Oncology, Hospital Universitario de Salamanca, Salamanca, Spain
| | - Reyes Bernabé
- Medical Oncology, Hospital U. Virgen Rocio, Seville, Spain
| | | | | | | | - Belén Sierra-Rodero
- Medical Oncology, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDIPHISA), Hospital Universitario Puerta de Hierro-Majadahonda, Majadahonda-Madrid, Spain
| | - Cristina Martinez-Toledo
- Medical Oncology, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDIPHISA), Hospital Universitario Puerta de Hierro-Majadahonda, Majadahonda-Madrid, Spain
| | - Ismael Fernández-Miranda
- Medical Oncology, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDIPHISA), Hospital Universitario Puerta de Hierro-Majadahonda, Majadahonda-Madrid, Spain
| | - Roberto Serna-Blanco
- Medical Oncology, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDIPHISA), Hospital Universitario Puerta de Hierro-Majadahonda, Majadahonda-Madrid, Spain
| | - Atocha Romero
- Medical Oncology, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDIPHISA), Hospital Universitario Puerta de Hierro-Majadahonda, Majadahonda-Madrid, Spain
| | - Virginia Calvo
- Medical Oncology, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDIPHISA), Hospital Universitario Puerta de Hierro-Majadahonda, Majadahonda-Madrid, Spain
| | - Alberto Cruz-Bermúdez
- Medical Oncology, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDIPHISA), Hospital Universitario Puerta de Hierro-Majadahonda, Majadahonda-Madrid, Spain
| |
Collapse
|
26
|
PD-1/PD-L1 inhibitors in treatment-naïve, advanced non-small cell lung cancer patients with < 1% PD-L1 expression: a meta-analysis of randomized controlled trials. J Cancer Res Clin Oncol 2022; 149:2179-2189. [PMID: 35984491 DOI: 10.1007/s00432-022-04286-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/13/2022] [Indexed: 10/15/2022]
Abstract
BACKGROUND PD-1/PD-L1 inhibitors prolong survival in treatment-naïve, locally advanced, and metastatic non-small cell lung cancer (NSCLC) with positive PD-L1 expression (> 1%/ > 50%). Recent evidence has suggested that tumors with < 1% PD-L1 expression may also be predictive of PD-1/PD-L1 inhibiting agents. METHODS Systematic review and meta-analysis were conducted of randomized controlled trials (RCTs) evaluating PD-1/PD-L1 inhibitors that have assessed tumors with < 1% PD-L1 expression (negative PD-L1 expression). PD-1/PD-L1 inhibitors-chemotherapy combinations (PC) were compared with histology-selected chemotherapy with respect to overall survival (OS) and progression-free survival (PFS). RESULTS Twelve RCTs comprising 5410 participants (PD-1/PD-L1 inhibitors-chemotherapy: 3051; chemotherapy: 2359) met the inclusion criteria. Tumors with PD-L1 expression < 1% were evident in 38.9% of the pooled study population. A significant OS [hazard ratio (HR) 0.71 95% confidence interval (CI) 0.63-0.80, p < 0.00001] and PFS [HR 0.65 95% CI 0.58-0.72, p < 0.00001] benefit of PC was evident in tumors with negative PD-L1 expression. PD-1/PD-L1 inhibitors-chemotherapy combinations were more likely to achieve an objective response than chemotherapy [odds ratio, 1.86; 95% CI, 1.46-2.38, p < 0.00001]. Histologic subtypes and diagnostic assays did not modify the OS and PFS treatment effects for PC compared to chemotherapy. CONCLUSION Tumors harboring < 1% PD-L1 expression are likely to benefit from PD-1/PD-L1 inhibitor-chemotherapy regimens in advanced NSCLC.
Collapse
|
27
|
Shi C, Wang Y, Xue J, Zhou X. Immunotherapy for EGFR-mutant advanced non-small-cell lung cancer: Current status, possible mechanisms and application prospects. Front Immunol 2022; 13:940288. [PMID: 35935943 PMCID: PMC9353115 DOI: 10.3389/fimmu.2022.940288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) are effective against advanced and even perioperative non-small-cell lung cancer (NSCLC) and result in durable clinical benefit, regardless of programmed death ligand-1 (PD-L1) expression status in cancer. Existing clinical evidence shows that the effect of immunotherapy in patients with EGFR-mutant NSCLC after the development of tyrosine kinase inhibitor (TKI) resistance is not satisfactory. However, compared with monotherapy, ICIs combined with chemotherapy can improve the efficacy. Encouragingly, compared with that of patients with sensitive mutations, the progression-free survival of patients with rare mutations who were treated with ICIs was increased. Adequately maximizing the efficacy of ICIs in EGFR-mutant NSCLC patients is worth exploring. In this review, we described preclinical and clinical studies of ICIs or combined therapy for EGFR-mutant NSCLC. We further focused on EGFR mutations and the cancer immune response, with particular attention given to the role of EGFR activation in the cancer-immunity cycle. The mechanisms for the natural resistance to ICIs were explored to identify corresponding countermeasures that made more EGFR-mutant NSCLC patients benefit from ICIs.
Collapse
Affiliation(s)
- Chunyan Shi
- Department of Thoracic Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- The Department of Oncology, Jiujiang No.1 People’s Hospital, Jiujiang, China
| | - Yan Wang
- Department of Thoracic Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jianxin Xue
- Department of Thoracic Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaojuan Zhou
- Department of Thoracic Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Xiaojuan Zhou,
| |
Collapse
|
28
|
Rojas F, Parra ER, Wistuba II, Haymaker C, Solis Soto LM. Pathological Response and Immune Biomarker Assessment in Non-Small-Cell Lung Carcinoma Receiving Neoadjuvant Immune Checkpoint Inhibitors. Cancers (Basel) 2022; 14:cancers14112775. [PMID: 35681755 PMCID: PMC9179283 DOI: 10.3390/cancers14112775] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 05/27/2022] [Accepted: 05/28/2022] [Indexed: 02/05/2023] Open
Abstract
Simple Summary Recently, the U.S. Food and Drug Administration (FDA) approved neoadjuvant immunotherapy plus chemotherapy for the treatment of resectable non-small-cell lung carcinoma (NSCLC) due to the clinical benefits reported in several clinical trials. In these settings, the pathological assessment of the tumor bed to quantify a pathological response has been used as a surrogate method of clinical benefit to neoadjuvant therapy. In addition, several clinical trials are including the assessment of tissue-, blood-, or host-based biomarkers to predict therapy response and to monitor the response to neoadjuvant treatment. In this manuscript, we provide an overview of current recommendations for the evaluation of pathological response and describe potential biomarkers used in clinical trials of neoadjuvant immunotherapy in resectable NSCLC. Abstract Lung cancer is the leading cause of cancer incidence and mortality worldwide. Adjuvant and neoadjuvant chemotherapy have been used in the perioperative setting of non-small-cell carcinoma (NSCLC); however, the five-year survival rate only improves by about 5%. Neoadjuvant treatment with immune checkpoint inhibitors (ICIs) has become significant due to improved survival in advanced NSCLC patients treated with immunotherapy agents. The assessment of pathology response has been proposed as a surrogate indicator of the benefits of neaodjuvant therapy. An outline of recommendations has been published by the International Association for the Study of Lung Cancer (IASLC) for the evaluation of pathologic response (PR). However, recent studies indicate that evaluations of immune-related changes are distinct in surgical resected samples from patients treated with immunotherapy. Several clinical trials of neoadjuvant immunotherapy in resectable NSCLC have included the study of biomarkers that can predict the response of therapy and monitor the response to treatment. In this review, we provide relevant information on the current recommendations of the assessment of pathological responses in surgical resected NSCLC tumors treated with neoadjuvant immunotherapy, and we describe current and potential biomarkers to predict the benefits of neoadjuvant immunotherapy in patients with resectable NSCLC.
Collapse
|
29
|
Provencio M, Serna-Blasco R, Nadal E, Insa A, García-Campelo MR, Casal Rubio J, Dómine M, Majem M, Rodríguez-Abreu D, Martínez-Martí A, De Castro Carpeño J, Cobo M, López Vivanco G, Del Barco E, Bernabé Caro R, Viñolas N, Barneto Aranda I, Viteri S, Pereira E, Royuela A, Calvo V, Martín-López J, García-García F, Casarrubios M, Franco F, Sánchez-Herrero E, Massuti B, Cruz-Bermúdez A, Romero A. Overall Survival and Biomarker Analysis of Neoadjuvant Nivolumab Plus Chemotherapy in Operable Stage IIIA Non-Small-Cell Lung Cancer (NADIM phase II trial). J Clin Oncol 2022; 40:2924-2933. [PMID: 35576508 PMCID: PMC9426809 DOI: 10.1200/jco.21.02660] [Citation(s) in RCA: 173] [Impact Index Per Article: 86.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Neoadjuvant chemotherapy plus nivolumab has been shown to be effective in resectable non–small-cell lung cancer (NSCLC) in the NADIM trial (ClinicalTrials.gov identifier: NCT03081689). The 3-year overall survival (OS) and circulating tumor DNA (ctDNA) analysis have not been reported.
Collapse
Affiliation(s)
| | | | - Ernest Nadal
- Institut Català d'Oncologia, L'Hospitalet De Llobregat, Barcelona, Spain
| | - Amelia Insa
- Fundación INCLIVA, Hospital Clínico Universitario de Valencia, Valencia, Spain
| | | | | | - Manuel Dómine
- Hospital Universitario Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| | | | | | - Alex Martínez-Martí
- Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | | | - Manuel Cobo
- Hospital Universitario Regional de Malaga, Spain
| | | | | | | | | | | | - Santiago Viteri
- Instituto Oncológico Dr. Rosell. Hospital Universitario Quiron Dexeus, Grupo QuironSalud, Barcelona, Spain
| | | | - Ana Royuela
- Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Virginia Calvo
- Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | | | | | | | - Fernando Franco
- Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Estela Sánchez-Herrero
- Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain.,Atrys Health, Barcelona, Spain
| | | | | | - Atocha Romero
- Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| |
Collapse
|
30
|
Provencio M, Calvo V, Romero A, Spicer JD, Cruz-Bermúdez A. Treatment Sequencing in Resectable Lung Cancer: The Good and the Bad of Adjuvant Versus Neoadjuvant Therapy. Am Soc Clin Oncol Educ Book 2022; 42:1-18. [PMID: 35561296 DOI: 10.1200/edbk_358995] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The treatment scenario for patients with resectable non-small cell lung cancer has changed dramatically with the incorporation of immunotherapy. The introduction of immunotherapy into treatment algorithms has yielded improved clinical outcomes in several phase II and III trials in both adjuvant (Impower010 and PEARLS) and neoadjuvant settings (JHU/MSK, LCMC3, NEOSTAR, Columbia/MGH, NADIM, and CheckMate-816), leading to new U.S. Food and Drug Administration approvals in this sense. Different treatment options are now available for patients, making the optimal treatment scenario a matter of intense debate. In this review, we summarize the main results concerning treatment sequencing in resectable non-small cell lung cancer from the past 30 years in the preimmunotherapy era, focusing on recent advances after incorporation of immunotherapy. Finally, the utility of several parameters (PD-L1, tumor mutational burden, radiomics, circulating tumor DNA, T-cell receptor, and immune populations) as predictive biomarkers for therapy personalization is discussed.
Collapse
Affiliation(s)
- Mariano Provencio
- Department of Medical Oncology, Hospital Universitario Puerta de Hierro, Madrid, Spain
| | - Virginia Calvo
- Department of Medical Oncology, Hospital Universitario Puerta de Hierro, Madrid, Spain
| | - Atocha Romero
- Department of Medical Oncology, Hospital Universitario Puerta de Hierro, Madrid, Spain
| | - Jonathan D Spicer
- Division of Thoracic Surgery, McGill University Health Centre, Montréal, Quebec, Canada
| | - Alberto Cruz-Bermúdez
- Department of Medical Oncology, Hospital Universitario Puerta de Hierro, Madrid, Spain
| |
Collapse
|
31
|
Mensali N, Inderberg EM. Emerging Biomarkers for Immunotherapy in Glioblastoma. Cancers (Basel) 2022; 14:1940. [PMID: 35454848 PMCID: PMC9024739 DOI: 10.3390/cancers14081940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 04/02/2022] [Accepted: 04/09/2022] [Indexed: 12/02/2022] Open
Abstract
Immunotherapy has shown clinical benefits in several solid malignancies-in particular, melanoma and non-small cell lung cancer. However, in other solid tumours such as glioblastoma (GBM), the response to immunotherapy has been more variable, and except for anti-PD-1 for patients with microsatellite instable (MSI)+ cancers, no immunotherapy is currently approved for GBM patients. GBM is the most common and most aggressive brain cancer with a very poor prognosis and a median overall survival of 15 months. A few prognostic biomarkers have been identified and are used to some extent, but apart from MSI, no biomarkers are used for patient stratification for treatments other than the standard of care, which was established 15 years ago. Around 25% of new treatments investigated in GBM are immunotherapies. Recent studies indicate that the use of integrated and validated immune correlates predicting the response and guiding treatments could improve the efficacy of immunotherapy in GBM. In this review, we will give an overview of the current status of immunotherapy and biomarkers in use in GBM with the main challenges of treatment in this disease. We will also discuss emerging biomarkers that could be used in future immunotherapy strategies for patient stratification and potentially improved treatment efficacy.
Collapse
Affiliation(s)
| | - Else Marit Inderberg
- Translational Research Unit, Department of Cellular Therapy, Oslo University Hospital, 0379 Oslo, Norway;
| |
Collapse
|
32
|
Aoki H, Shichino S, Matsushima K, Ueha S. Revealing Clonal Responses of Tumor-Reactive T-Cells Through T Cell Receptor Repertoire Analysis. Front Immunol 2022; 13:807696. [PMID: 35154125 PMCID: PMC8829044 DOI: 10.3389/fimmu.2022.807696] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 01/12/2022] [Indexed: 12/14/2022] Open
Abstract
CD8+ T cells are the key effector cells that contribute to the antitumor immune response. They comprise various T-cell clones with diverse antigen-specific T-cell receptors (TCRs). Thus, elucidating the overall antitumor responses of diverse T-cell clones is an emerging challenge in tumor immunology. With the recent advancement in next-generation DNA sequencers, comprehensive analysis of the collection of TCR genes (TCR repertoire analysis) is feasible and has been used to investigate the clonal responses of antitumor T cells. However, the immunopathological significance of TCR repertoire indices is still undefined. In this review, we introduce two approaches that facilitate an immunological interpretation of the TCR repertoire data: inter-organ clone tracking analysis and single-cell TCR sequencing. These approaches for TCR repertoire analysis will provide a more accurate understanding of the response of tumor-specific T cells in the tumor microenvironment.
Collapse
Affiliation(s)
- Hiroyasu Aoki
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan.,Department of Hygiene, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shigeyuki Shichino
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Kouji Matsushima
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Satoshi Ueha
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| |
Collapse
|
33
|
Sierra-Rodero B, Cruz-Bermúdez A, Nadal E, Garitaonaindía Y, Insa A, Mosquera J, Casal-Rubio J, Dómine M, Majem M, Rodriguez-Abreu D, Martinez-Marti A, De Castro Carpeño J, Cobo M, López Vivanco G, Del Barco E, Bernabé Caro R, Viñolas N, Barneto Aranda I, Viteri S, Massuti B, Laza-Briviesca R, Casarrubios M, García-Grande A, Romero A, Franco F, Provencio M. Clinical and molecular parameters associated to pneumonitis development in non-small-cell lung cancer patients receiving chemoimmunotherapy from NADIM trial. J Immunother Cancer 2021; 9:e002804. [PMID: 34446577 PMCID: PMC8395363 DOI: 10.1136/jitc-2021-002804] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2021] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Pneumonitis (Pn) is one of the main immune-related adverse effects, having a special importance in lung cancer, since they share affected tissue. Despite its clinical relevance, Pn development remains an unpredictable treatment adverse effect, whose mechanisms are mainly unknown, being even more obscure when it is associated to chemoimmunotherapy. METHODS In order to identify parameters associated to treatment related Pn, we analyzed clinical variables and molecular parameters from 46 patients with potentially resectable stage IIIA non-small-cell lung cancer treated with neoadjuvant chemoimmunotherapy included in the NADIM clinical trial (NCT03081689). Pn was defined as clinical or radiographic evidence of lung inflammation without alternative diagnoses, from treatment initiation to 180 days. RESULTS Among 46 patients, 12 developed Pn (26.1%). Sex, age, smoking status, packs-year, histological subtype, clinical or pathological response, progression-free survival, overall survival and number of nivolumab cycles, were not associated to Pn development. Regarding molecular parameters at diagnosis, Pn development was not associated to programmed death ligand 1, TPS, T cell receptor repertoire parameters, or tumor mutational burden. However, patients who developed Pn had statistically significant lower blood median levels of platelet to monocyte ratio (p=0.012) and teratocarcinoma-derived growth factor 1 (p=0.013; area under the curve (AUC) 0.801), but higher median percentages of natural killers (NKs) (p=0.019; AUC 0.786), monocytes (p=0.017; AUC 0.791), MSP (p=0.006; AUC 0.838), PARN (p=0.017; AUC 0.790), and E-Cadherin (p=0.022; AUC 0.788). In addition, the immune scenario of Pn after neoadjuvant treatment involves: high levels of neutrophils and NK cells, but low levels of B and T cells in peripheral blood; increased clonality of intratumoral T cells; and elevated plasma levels of several growth factors (EGF, HGF, VEGF, ANG-1, PDGF, NGF, and NT4) and inflammatory cytokines (MIF, CCL16, neutrophil gelatinase-associated lipocalin, BMP-4, and u-PAR). CONCLUSIONS Although statistically underpowered, our results shed light on the possible mechanisms behind Pn development, involving innate and adaptative immunity, and open the possibility to predict patients at high risk. If confirmed, this may allow the personalization of both, the surveillance strategy and the therapeutic approaches to manage Pn in patients receiving chemoimmunotherapy.
Collapse
Affiliation(s)
- Belén Sierra-Rodero
- Oncología Médica, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDIPHISA), Hospital Universitario Puerta de Hierro, Majadahonda, Madrid, Spain
| | - Alberto Cruz-Bermúdez
- Oncología Médica, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDIPHISA), Hospital Universitario Puerta de Hierro, Majadahonda, Madrid, Spain
| | - Ernest Nadal
- Department of Medical Oncology, Catalan Institute of Oncology, Oncobell Program, IDIBELL, L'Hospitalet de Llobregat, L'Hospitalet, Barcelona, Spain
| | - Yago Garitaonaindía
- Oncología Médica, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDIPHISA), Hospital Universitario Puerta de Hierro, Majadahonda, Madrid, Spain
| | - Amelia Insa
- Medical Oncology, INCLIVA, Valencia, Valencia, Spain
| | - Joaquín Mosquera
- Medical Oncology, Hospital Universitario A Coruña, A Coruña, Spain
| | | | - Manuel Dómine
- Medical Oncology, Hospital Universitario Fundación Jiménez Díaz, Madrid, Spain
| | - Margarita Majem
- Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona, Catalunya, Spain
| | - Delvys Rodriguez-Abreu
- Medical Oncology, Hospital Universitario Insular de Gran Canaria, Las Palmas, Canarias, Spain
| | | | | | - Manuel Cobo
- Medical Oncology, Hospital Regional Universitario de Málaga, Malaga, Andalucía, Spain
| | | | - Edel Del Barco
- Medical Oncology, Hospital Universitario de Salamanca, Salamanca, Spain
| | | | - Nuria Viñolas
- Medical Oncology, Hospital Clínic de Barcelona, Barcelona, Catalunya, Spain
| | | | - Santiago Viteri
- Instituto Oncológico Dr. Rosell. Hospital Universitario Quiron Dexeus, Barcelona, Spain
| | - Bartomeu Massuti
- Medical Oncology, Alicante General University Hospital, Alicante, Valencia, Spain
| | - Raquel Laza-Briviesca
- Oncología Médica, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDIPHISA), Hospital Universitario Puerta de Hierro, Majadahonda, Madrid, Spain
| | - Marta Casarrubios
- Oncología Médica, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDIPHISA), Hospital Universitario Puerta de Hierro, Majadahonda, Madrid, Spain
| | - Aránzazu García-Grande
- Flow Cytometry Core Facility, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDIPHISA), Majadahonda, Spain
| | - Atocha Romero
- Oncología Médica, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDIPHISA), Hospital Universitario Puerta de Hierro, Majadahonda, Madrid, Spain
| | - Fernando Franco
- Oncología Médica, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDIPHISA), Hospital Universitario Puerta de Hierro, Majadahonda, Madrid, Spain
| | - Mariano Provencio
- Oncología Médica, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDIPHISA), Hospital Universitario Puerta de Hierro, Majadahonda, Madrid, Spain
| |
Collapse
|