401
|
Sachpekidis C, Stein-Thoeringer CK, Kopp-Schneider A, Weru V, Dimitrakopoulou-Strauss A, Hassel JC. Can physiologic colonic [ 18F]FDG uptake in PET/CT imaging predict response to immunotherapy in metastatic melanoma? Eur J Nucl Med Mol Imaging 2023; 50:3709-3722. [PMID: 37452874 PMCID: PMC10547632 DOI: 10.1007/s00259-023-06327-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 06/27/2023] [Indexed: 07/18/2023]
Abstract
AIM The development of biomarkers that can reliably and early predict response to immune checkpoint inhibitors (ICIs) is crucial in melanoma. In recent years, the gut microbiome has emerged as an important regulator of immunotherapy response, which may, moreover, serve as a surrogate marker and prognosticator in oncological patients under immunotherapy. Aim of the present study is to investigate if physiologic colonic [18F]FDG uptake in PET/CT before start of ICIs correlates with clinical outcome of metastatic melanoma patients. The relation between [18F]FDG uptake in lymphoid cell-rich organs and long-term patient outcome is also assessed. METHODOLOGY One hundred nineteen stage IV melanoma patients scheduled for immunotherapy with ipilimumab, applied either as monotherapy or in combination with nivolumab, underwent baseline [18F]FDG PET/CT. PET/CT data analysis consisted of standardized uptake value (SUV), metabolic tumor volume (MTV), and total lesion glycolysis (TLG) calculations in the colon as well as measurements of the colon-to-liver SUV ratios (CLRmean, CLRmax). Visual grading of colon uptake based on a four-point scale was also performed. Moreover, the spleen-to-liver SUV ratios (SLRmean, SLRmax) and the bone marrow-to-liver SUV ratios (BLRmean, BLRmax) were calculated. We also measured serum lipopolysaccharide (LPS) levels as a marker for bacterial translocation and surrogate for mucosal defense homeostasis. The results were correlated with patients' best clinical response, progression-free survival (PFS), and overall survival (OS) as well as clinical signs of colitis. RESULTS Median follow-up [95%CI] from the beginning of immunotherapy was 64.6 months [61.0-68.6 months]. Best response to treatment was progressive disease (PD) for 60 patients, stable disease (SD) for 37 patients, partial response (PR) for 18 patients, and complete response (CR) for 4 patients. Kaplan-Meier curves demonstrated a trend for longer PFS and OS in patients with lower colonic SUV and CLR values; however, no statistical significance for these parameters as prognostic factors was demonstrated. On the other hand, patients showing disease control as best response to treatment (SD, PR, CR) had significantly lower colonic MTV and TLG than those showing PD. With regard to lymphoid cell-rich organs, significantly lower baseline SLRmax and BLRmax were observed in patients responding with disease control than progression to treatment. Furthermore, patients with lower SLRmax and BLRmax values had a significantly longer OS when dichotomized at their median. In multivariate analysis, PET parameters that were found to significantly adversely correlate with patient survival were colonic MTV for PFS, colonic TLG for PFS, and BLRmax for PFS and OS. CONCLUSIONS Physiologic colonic [18F]FDG uptake in PET/CT, as assessed by means of SUV, before start of ipilimumab-based treatment does not seem to independently predict patient survival of metastatic melanoma. On the other hand, volumetric PET parameters, such as MTV and TLG, derived from the normal gut may identify patients showing disease control to immunotherapy and significantly correlate with PFS. Moreover, the investigation of glucose metabolism in the spleen and the bone marrow may offer prognostic information.
Collapse
Affiliation(s)
- Christos Sachpekidis
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69210, Heidelberg, Germany.
| | - Christoph K Stein-Thoeringer
- Laboratory of Translational, Microbiome Science, Internal Medicine I, University Clinic Tuebingen, Tuebingen, Germany
| | | | - Vivienn Weru
- Department of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Antonia Dimitrakopoulou-Strauss
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69210, Heidelberg, Germany
| | - Jessica C Hassel
- Department of Dermatology and National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
402
|
Pavlick AC, Ariyan CE, Buchbinder EI, Davar D, Gibney GT, Hamid O, Hieken TJ, Izar B, Johnson DB, Kulkarni RP, Luke JJ, Mitchell TC, Mooradian MJ, Rubin KM, Salama AK, Shirai K, Taube JM, Tawbi HA, Tolley JK, Valdueza C, Weiss SA, Wong MK, Sullivan RJ. Society for Immunotherapy of Cancer (SITC) clinical practice guideline on immunotherapy for the treatment of melanoma, version 3.0. J Immunother Cancer 2023; 11:e006947. [PMID: 37852736 PMCID: PMC10603365 DOI: 10.1136/jitc-2023-006947] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2023] [Indexed: 10/20/2023] Open
Abstract
Since the first approval for immune checkpoint inhibitors (ICIs) for the treatment of cutaneous melanoma more than a decade ago, immunotherapy has completely transformed the treatment landscape of this chemotherapy-resistant disease. Combination regimens including ICIs directed against programmed cell death protein 1 (PD-1) with anti-cytotoxic T lymphocyte antigen-4 (CTLA-4) agents or, more recently, anti-lymphocyte-activation gene 3 (LAG-3) agents, have gained regulatory approvals for the treatment of metastatic cutaneous melanoma, with long-term follow-up data suggesting the possibility of cure for some patients with advanced disease. In the resectable setting, adjuvant ICIs prolong recurrence-free survival, and neoadjuvant strategies are an active area of investigation. Other immunotherapy strategies, such as oncolytic virotherapy for injectable cutaneous melanoma and bispecific T-cell engager therapy for HLA-A*02:01 genotype-positive uveal melanoma, are also available to patients. Despite the remarkable efficacy of these regimens for many patients with cutaneous melanoma, traditional immunotherapy biomarkers (ie, programmed death-ligand 1 expression, tumor mutational burden, T-cell infiltrate and/or microsatellite stability) have failed to reliably predict response. Furthermore, ICIs are associated with unique toxicity profiles, particularly for the highly active combination of anti-PD-1 plus anti-CTLA-4 agents. The Society for Immunotherapy of Cancer (SITC) convened a panel of experts to develop this clinical practice guideline on immunotherapy for the treatment of melanoma, including rare subtypes of the disease (eg, uveal, mucosal), with the goal of improving patient care by providing guidance to the oncology community. Drawing from published data and clinical experience, the Expert Panel developed evidence- and consensus-based recommendations for healthcare professionals using immunotherapy to treat melanoma, with topics including therapy selection in the advanced and perioperative settings, intratumoral immunotherapy, when to use immunotherapy for patients with BRAFV600-mutated disease, management of patients with brain metastases, evaluation of treatment response, special patient populations, patient education, quality of life, and survivorship, among others.
Collapse
Affiliation(s)
| | - Charlotte E Ariyan
- Department of Surgery Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | - Diwakar Davar
- Hillman Cancer Center, University of Pittsburg Medical Center, Pittsburgh, Pennsylvania, USA
| | - Geoffrey T Gibney
- Lombardi Comprehensive Cancer Center, MedStar Georgetown University Hospital, Washington, District of Columbia, USA
| | - Omid Hamid
- The Angeles Clinic and Research Institute, A Cedars-Sinai Affiliate, Los Angeles, California, USA
| | - Tina J Hieken
- Department of Surgery and Comprehensive Cancer Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Benjamin Izar
- Department of Medicine, Division of Hematology/Oncology, Columbia University Medical Center, New York, New York, USA
| | - Douglas B Johnson
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Rajan P Kulkarni
- Departments of Dermatology, Oncological Sciences, Biomedical Engineering, and Center for Cancer Early Detection Advanced Research, Knight Cancer Institute, OHSU, Portland, Oregon, USA
- Operative Care Division, VA Portland Health Care System (VAPORHCS), Portland, Oregon, USA
| | - Jason J Luke
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Tara C Mitchell
- Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Meghan J Mooradian
- Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Krista M Rubin
- Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - April Ks Salama
- Department of Medicine, Division of Medical Oncology, Duke University, Durham, Carolina, USA
| | - Keisuke Shirai
- Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Janis M Taube
- Department of Dermatology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Hussein A Tawbi
- Department of Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - J Keith Tolley
- Patient Advocate, Melanoma Research Alliance, Washington, DC, USA
| | - Caressa Valdueza
- Cutaneous Oncology Program, Weill Cornell Medicine, New York, New York, USA
| | - Sarah A Weiss
- Department of Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
| | - Michael K Wong
- Patient Advocate, Melanoma Research Alliance, Washington, DC, USA
| | - Ryan J Sullivan
- Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
403
|
Placke JM, Kimmig M, Griewank K, Herbst R, Terheyden P, Utikal J, Pföhler C, Ulrich J, Kreuter A, Mohr P, Gutzmer R, Meier F, Dippel E, Welzel J, Engel DR, Kreft S, Sucker A, Lodde G, Krefting F, Stoffels I, Klode J, Roesch A, Zimmer L, Livingstone E, Hadaschik E, Becker JC, Weichenthal M, Tasdogan A, Schadendorf D, Ugurel S. Correlation of tumor PD-L1 expression in different tissue types and outcome of PD-1-based immunotherapy in metastatic melanoma - analysis of the DeCOG prospective multicenter cohort study ADOREG/TRIM. EBioMedicine 2023; 96:104774. [PMID: 37660535 PMCID: PMC10483509 DOI: 10.1016/j.ebiom.2023.104774] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 08/10/2023] [Accepted: 08/10/2023] [Indexed: 09/05/2023] Open
Abstract
BACKGROUND PD-1-based immune checkpoint inhibition (ICI) is the major backbone of current melanoma therapy. Tumor PD-L1 expression represents one of few biomarkers predicting ICI therapy outcome. The objective of the present study was to systematically investigate whether the type of tumor tissue examined for PD-L1 expression has an impact on the correlation with ICI therapy outcome. METHODS Pre-treatment tumor tissue was collected within the prospective DeCOG cohort study ADOREG/TRIM (CA209-578; NCT05750511) between February 2014 and May 2020 from 448 consecutive patients who received PD-1-based ICI for non-resectable metastatic melanoma. The primary study endpoint was best overall response (BOR), secondary endpoints were progression-free (PFS) and overall survival (OS). All endpoints were correlated with tumor PD-L1 expression (quantified with clone 28-8; cutoff ≥5%) and stratified by tissue type. FINDINGS Tumor PD-L1 was determined in 95 primary tumors (PT; 36.8% positivity), 153 skin/subcutaneous (34.0% positivity), 115 lymph node (LN; 50.4% positivity), and 85 organ (40.8% positivity) metastases. Tumor PD-L1 correlated with BOR if determined in LN (OR = 0.319; 95% CI = 0.138-0.762; P = 0.010), but not in skin/subcutaneous metastases (OR = 0.656; 95% CI = 0.311-1.341; P = 0.26). PD-L1 positivity determined on LN metastases was associated with favorable survival (PFS, HR = 0.490; 95% CI = 0.310-0.775; P = 0.002; OS, HR = 0.519; 95% CI = 0.307-0.880; P = 0.014). PD-L1 positivity determined in PT (PFS, HR = 0.757; 95% CI = 0.467-1.226; P = 0.27; OS; HR = 0.528; 95% CI = 0.305-0.913; P = 0.032) was correlated with survival to a lesser extent. No relevant survival differences were detected by PD-L1 determined in skin/subcutaneous metastases (PFS, HR = 0.825; 95% CI = 0.555-1.226; P = 0.35; OS, HR = 1.083; 95% CI = 0.698-1.681; P = 0.72). INTERPRETATION For PD-1-based immunotherapy in melanoma, tumor PD-L1 determined in LN metastases was stronger correlated with therapy outcome than that assessed in PT or organ metastases. PD-L1 determined in skin/subcutaneous metastases showed no outcome correlation and therefore should be used with caution for clinical decision making. FUNDING Bristol-Myers Squibb (ADOREG/TRIM, NCT05750511); German Research Foundation (DFG; Clinician Scientist Program UMEA); Else Kröner-Fresenius-Stiftung (EKFS; Medical Scientist Academy UMESciA).
Collapse
Affiliation(s)
- Jan-Malte Placke
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany; German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Germany.
| | - Mona Kimmig
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany.
| | - Klaus Griewank
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany.
| | | | | | - Jochen Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany; DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany.
| | - Claudia Pföhler
- Department of Dermatology, Saarland University Medical School, Homburg/Saar, Germany.
| | - Jens Ulrich
- Skin Cancer Center, Department of Dermatology, Harz Clinics, Quedlinburg, Germany.
| | - Alexander Kreuter
- Department of Dermatology, Venereology and Allergology, Helios St. Elisabeth Klinik Oberhausen, University Witten-Herdecke, Oberhausen, Germany.
| | - Peter Mohr
- Department of Dermatology, Elbe Kliniken Buxtehude, Buxtehude, Germany.
| | - Ralf Gutzmer
- Department of Dermatology, Skin Cancer Center Minden, Minden, Germany.
| | - Friedegund Meier
- Department of Dermatology, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany.
| | - Edgar Dippel
- Department of Dermatology, Ludwigshafen Medical Center, Ludwigshafen, Germany.
| | - Julia Welzel
- Department of Dermatology, Augsburg Medical Center, Augsburg, Germany.
| | - Daniel Robert Engel
- Department of Immunodynamics, Institute for Experimental Immunology and Imaging, Medical Research Centre, University Hospital Essen, Essen, Germany; German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Germany.
| | - Sophia Kreft
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany.
| | - Antje Sucker
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany.
| | - Georg Lodde
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany.
| | - Frederik Krefting
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany.
| | - Ingo Stoffels
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany.
| | - Joachim Klode
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany.
| | - Alexander Roesch
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany; German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Germany.
| | - Lisa Zimmer
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany.
| | - Elisabeth Livingstone
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany.
| | - Eva Hadaschik
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany.
| | - Jürgen C Becker
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany; Translational Skin Cancer Research, West German Cancer Center, University Medicine Essen, Essen, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Germany.
| | - Michael Weichenthal
- Department of Dermatology, University Hospital of Schleswig-Holstein, Kiel, Germany.
| | - Alpaslan Tasdogan
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany; German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Germany.
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany; German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Germany.
| | - Selma Ugurel
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany; German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Germany.
| |
Collapse
|
404
|
Mor E, Schtrechman G, Nizri E, Shimonovitz M, Asher N, Ben-Betzalel G, Grynberg S, Stoff R, Miodovnik M, Adileh M, Ben-Yaacov A, Steinberg Y, Shapira R, Schachter J, Lahat G, Nissan A, Zippel D, Laks S. PET-CT underestimates the true pathological extent of disease at lymphadenectomy for melanoma patients after systemic therapy. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2023; 49:106950. [PMID: 37301639 DOI: 10.1016/j.ejso.2023.06.002] [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: 01/28/2023] [Revised: 05/20/2023] [Accepted: 06/05/2023] [Indexed: 06/12/2023]
Abstract
INTRODUCTION Modern systemic therapy has revolutionized the treatment of melanoma. Currently, patients with clinically involved lymph nodes require lymphadenectomy with associated morbidities. Positron Emission Tomography - Computed Tomography (PET-CT) has demonstrated accuracy in melanoma detection and response to therapy. We aimed to identify whether a PET-CT directed lymphatic resection after systemic therapy is oncologically sound. MATERIALS AND METHODS Retrospective review of patients who underwent lymphadenectomy after systemic therapy for melanoma with a preoperative PET-CT. Examined demographic, clinical, and perioperative parameters including extent of disease, systemic therapy and response, and PET-CT findings compared to pathological outcomes. We compared patients with "as or less than expected" outcomes on pathology against those with "more than expected" pathological outcomes. RESULTS Thirty-nine patients met inclusion criteria. In 28 (71.8%), pathological outcomes were "as or less than expected" by PET-CT, and in 11 (28.2%) pathological outcome were "more than expected". "More than expected" occurred more frequently with advanced disease at presentation with 75% presenting with regional/metastatic disease versus only 42.9% in the "as or less than expected" group (p = 0.015). Poor response to therapy also trended towards the "more than expected" group with only 27.3% favorable response versus 53.6% favorable response in the "as or less than expected" group, not statistically significant. Extent of disease on imaging failed to predict pathological concordance. CONCLUSION PET-CT underestimates pathological extent of disease in the lymphatic basin in 30% of patients after systemic therapy. We failed to identify predictors of more extensive disease and warn against limited PET-CT directed lymphatic resections.
Collapse
Affiliation(s)
- Eyal Mor
- Sheba Tel Hashomer Medical Center, Department of Surgery C and Surgical Oncology, Ramat Gan, Israel
| | - Gal Schtrechman
- Sheba Tel Hashomer Medical Center, Department of Surgery C and Surgical Oncology, Ramat Gan, Israel
| | - Eran Nizri
- Department of Surgery B, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Michal Shimonovitz
- Department of Surgery B, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Nethanel Asher
- Sheba Tel Hashomer Medical Center, Department of Oncology, Ramat Gan, Israel
| | - Guy Ben-Betzalel
- Sheba Tel Hashomer Medical Center, Department of Oncology, Ramat Gan, Israel
| | - Shirly Grynberg
- Sheba Tel Hashomer Medical Center, Department of Oncology, Ramat Gan, Israel
| | - Ronen Stoff
- Sheba Tel Hashomer Medical Center, Department of Oncology, Ramat Gan, Israel
| | - Mor Miodovnik
- Institute of Oncology, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Mohammad Adileh
- Sheba Tel Hashomer Medical Center, Department of Surgery C and Surgical Oncology, Ramat Gan, Israel
| | - Almog Ben-Yaacov
- Sheba Tel Hashomer Medical Center, Department of Surgery C and Surgical Oncology, Ramat Gan, Israel
| | - Yael Steinberg
- Sheba Tel Hashomer Medical Center, Department of Oncology, Ramat Gan, Israel
| | - Ronnie Shapira
- Sheba Tel Hashomer Medical Center, Department of Oncology, Ramat Gan, Israel
| | - Jacob Schachter
- Sheba Tel Hashomer Medical Center, Department of Oncology, Ramat Gan, Israel
| | - Guy Lahat
- Department of Surgery B, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Aviram Nissan
- Sheba Tel Hashomer Medical Center, Department of Surgery C and Surgical Oncology, Ramat Gan, Israel
| | - Douglas Zippel
- Sheba Tel Hashomer Medical Center, Department of Surgery C and Surgical Oncology, Ramat Gan, Israel
| | - Shachar Laks
- Sheba Tel Hashomer Medical Center, Department of Surgery C and Surgical Oncology, Ramat Gan, Israel.
| |
Collapse
|
405
|
Yuan P, Li S, Xiong S, Zheng F, Yang L, Jiang M, Liu J, Liu X, Deng J, Zeng J, Fu B. Development of a butyrate metabolism-related gene-based molecular subtypes and scoring system for predicting prognosis and immunotherapy response in bladder cancer. J Cancer Res Clin Oncol 2023; 149:12489-12505. [PMID: 37450031 DOI: 10.1007/s00432-023-05067-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 06/29/2023] [Indexed: 07/18/2023]
Abstract
PURPOSE In recent times, multiple molecular subtypes with varying prognoses have been identified in bladder cancer (BLCA). However, the attributes of butyrate metabolism-related (BMR) molecular subtypes and their correlation with immunotherapy response remain inadequately explored in BLCA. METHODS We utilized 594 samples of BLCA to investigate the molecular subtypes mediated by BMR genes and their correlation with the immunotherapy response. To quantify the BMR features of individual tumors, we developed a BMR score through the COX and LASSO regression methods. Clinical-related, tumor microenvironment, drug-sensitive and immunotherapy analyses were used to comprehensively analyze BMR scores. RESULTS Two distinct molecular subtypes related to butyrate metabolism were identified in BLCA, each with unique prognostic implications and immune microenvironments. BMR score was constructed based on 7 BMR genes and was used to classify the patients into two score groups. Clinical analysis revealed that the BMR score was an independent prognostic factor. The higher the score, the worse the prognosis. The BMR score can also predict tumor immunity. The results demonstrated that a low BMR score was associated with higher efficacy of immunotherapy, which was also validated by an external dataset. CONCLUSION Our study proposes both molecular subtypes and a BMR-based score as promising prognostic classifications in BLCA. These findings may offer new insights for the development of precise targeted cancer therapies.
Collapse
Affiliation(s)
- Peng Yuan
- Department of Ophthalmology, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, China
- Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, China
| | - Sheng Li
- Department of Urology, Nanchang University, Nanchang, China
- The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Situ Xiong
- Department of Urology, Nanchang University, Nanchang, China
- The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Fucun Zheng
- Department of Urology, Nanchang University, Nanchang, China
- The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Lin Yang
- Department of Urology, Nanchang University, Nanchang, China
- The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ming Jiang
- Department of Urology, Nanchang University, Nanchang, China
- The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jiahao Liu
- Department of Urology, Nanchang University, Nanchang, China
- The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiaoqiang Liu
- Department of Urology, Nanchang University, Nanchang, China.
- The First Affiliated Hospital of Nanchang University, Nanchang, China.
| | - Jun Deng
- Department of Urology, Nanchang University, Nanchang, China
- The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jin Zeng
- Department of Urology, Nanchang University, Nanchang, China.
- The First Affiliated Hospital of Nanchang University, Nanchang, China.
| | - Bin Fu
- Department of Urology, Nanchang University, Nanchang, China.
- The First Affiliated Hospital of Nanchang University, Nanchang, China.
| |
Collapse
|
406
|
Zhang M, Liu J, Xia Q. Role of gut microbiome in cancer immunotherapy: from predictive biomarker to therapeutic target. Exp Hematol Oncol 2023; 12:84. [PMID: 37770953 PMCID: PMC10537950 DOI: 10.1186/s40164-023-00442-x] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 09/04/2023] [Indexed: 09/30/2023] Open
Abstract
Immunotherapy has emerged as an effective treatment for various types of cancers. Recent studies have highlighted a significant correlation between the gut microbiome and patients' response to immunotherapy. Several characteristics of the gut microbiome, such as community structures, taxonomic compositions, and molecular functions, have been identified as crucial biomarkers for predicting immunotherapy response and immune-related adverse events (irAEs). Unlike other -omics, the gut microbiome can serve as not only biomarkers but also potential targets for enhancing the efficacy of immunotherapy. Approaches for modulating the gut microbiome include probiotics/prebiotics supplementation, dietary interventions, fecal microbiota transplantation (FMT), and antibiotic administration. This review primarily focuses on elucidating the potential role of the gut microbiome in predicting the response to cancer immunotherapy and improving its efficacy. Notably, we explore reasons behind inconsistent findings observed in different studies, and highlight the underlying benefits of antibiotics in liver cancer immunotherapy.
Collapse
Affiliation(s)
- Mengwei Zhang
- Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai, China
- Shanghai Institute of Transplantation, Shanghai, China
| | - Jinkai Liu
- Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai, China
- Shanghai Institute of Transplantation, Shanghai, China
| | - Qiang Xia
- Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai, China
- Shanghai Institute of Transplantation, Shanghai, China
| |
Collapse
|
407
|
Sauer N, Szlasa W, Szewczyk A, Novickij V, Saczko J, Baczyńska D, Daczewska M, Kulbacka J. Effects of Nanosecond Pulsed Electric Field on Immune Checkpoint Receptors in Melanoma Cells. Pharmaceuticals (Basel) 2023; 16:1362. [PMID: 37895833 PMCID: PMC10610193 DOI: 10.3390/ph16101362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/11/2023] [Accepted: 09/20/2023] [Indexed: 10/29/2023] Open
Abstract
Checkpoint molecules such as PD-1, LAG-3, and TIM-3 are currently under extensive investigation for their roles in the attenuation of the immune response in cancer. Various methods have been applied to overcome the challenges in this field. This study investigated the effects of nanosecond pulsed electric field (nsPEF) treatment on the expression of immune checkpoint molecules in A375 and C32 melanoma cells. The researchers found that the nsPEF treatment was able to enhance membrane permeabilization and morphological changes in the cell membrane without being cytotoxic. We found that the effects of nsPEFs on melanoma included (1) the transport of vesicles from the inside to the outside of the cells, (2) cell contraction, and (3) the migration of lipids from inside the cells to their peripheries. The treatment increased the expression of PD-1 checkpoint receptors. Furthermore, we also observed potential co-localization or clustering of MHC class II and PD-1 molecules on the cell surface and the secretion of cytokines such as TNF-α and IL-6. These findings suggest that nsPEF treatment could be a viable approach to enhance the delivery of therapeutic agents to cancer cells and to modulate the tumor microenvironment to promote an antitumor immune response. Further studies are needed to explore the mechanisms underlying these effects and their impacts on the antitumor immune response, and to investigate the potential of nsPEF treatment in combination with immune checkpoint inhibitors to improve clinical outcomes for cancer patients.
Collapse
Affiliation(s)
- Natalia Sauer
- Faculty of Pharmacy, Wroclaw Medical University, 50-556 Wroclaw, Poland;
| | - Wojciech Szlasa
- Faculty of Medicine, Wroclaw Medical University, 50-556 Wroclaw, Poland;
| | - Anna Szewczyk
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, 51-618 Wroclaw, Poland; (A.S.); (D.B.)
- Department of Animal Developmental Biology, Faculty of Biological Sciences, University of Wroclaw, Sienkiewicza 21, 50-335 Wroclaw, Poland;
| | - Vitalij Novickij
- Institute of High Magnetic Fields, Vilnius Gediminas Technical University, 08217 Vilnius, Lithuania;
- Department of Immunology, State Research Institute Centre for Innovative Medicine, Santariškių 5, 08410 Vilnius, Lithuania
| | - Jolanta Saczko
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, 51-618 Wroclaw, Poland; (A.S.); (D.B.)
| | - Dagmara Baczyńska
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, 51-618 Wroclaw, Poland; (A.S.); (D.B.)
| | - Małgorzata Daczewska
- Department of Animal Developmental Biology, Faculty of Biological Sciences, University of Wroclaw, Sienkiewicza 21, 50-335 Wroclaw, Poland;
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, 51-618 Wroclaw, Poland; (A.S.); (D.B.)
- Department of Immunology, State Research Institute Centre for Innovative Medicine, Santariškių 5, 08410 Vilnius, Lithuania
| |
Collapse
|
408
|
Xu M, Li G, Li J, Xiong H, He S. Pharmacovigilance for rare diseases: a bibliometrics and knowledge-map analysis based on web of science. Orphanet J Rare Dis 2023; 18:303. [PMID: 37752556 PMCID: PMC10523788 DOI: 10.1186/s13023-023-02915-y] [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: 05/31/2023] [Accepted: 09/07/2023] [Indexed: 09/28/2023] Open
Abstract
OBJECTIVES The aims of this paper is to search and explore publications in the field of pharmacovigilance for rare diseases and to visualize general information, research hotspots, frontiers and future trends in the field using the bibliometric tool CiteSpace to provide evidence-based evidence for scholars. METHODS We searched the Web of Science Core Collection (WoSCC) for studies related to pharmacovigilance for rare diseases, spanning January 1, 1997-October 25, 2022. CiteSpace software was utilized to discuss countries/regions, institutions, authors, journals, and keywords. RESULTS After screening, a total of 599 valid publications were included in this study, with a significant upward trend in the number of publications. These studies were from 68 countries/regions with the United States and the United Kingdom making the largest contributions to the field. 4,806 research scholars from 493 institutions conducted studies on pharmacovigilance for rare diseases. Harvard University and University of California were the top two productive institutions in the research field. He Dian of the Affiliated Hospital of Guizhou Medical University and Peter G.M. Mol of the University of Groningen, The Netherlands, were the two most prolific researchers. The Cochrane Database of Systematic Reviews and the New England Journal of Medicine were the journals with the highest number of articles and co-citation frequency respectively. Clinical trial, therapy and adverse event were the top three most cited keywords. CONCLUSIONS Based on keywords co-occurrence analysis, four research topics were identified: orphan drug clinical trials, postmarketing ADR surveillance for orphan drugs, rare diseases and orphan drug management, and diagnosis and treatment of rare diseases. Immune-related adverse reactions and benefit-risk assessment of enzyme replacement therapy were at the forefront of research in this field. Treatment outcomes, early diagnosis and natural history studies of rare diseases may become hotspots for future research.
Collapse
Affiliation(s)
- Mengdan Xu
- School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China.
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangzhou, Guangdong, China.
| | - Guozhi Li
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangzhou, Guangdong, China
| | - Jiazhao Li
- School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Huiyu Xiong
- Center for ADR Monitoring of Guangdong, Guangzhou, Guangdong, China
| | - Suzhen He
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
409
|
Tangudu NK, Buj R, Wang H, Wang J, Cole AR, Uboveja A, Fang R, Amalric A, Sajjakulnukit P, Lyons MA, Cooper K, Hempel N, Snyder NW, Lyssiotis CA, Chandran UR, Aird KM. De novo purine metabolism is a metabolic vulnerability of cancers with low p16 expression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.15.549149. [PMID: 37503050 PMCID: PMC10369956 DOI: 10.1101/2023.07.15.549149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
p16 is a tumor suppressor encoded by the CDKN2A gene whose expression is lost in ~50% of all human cancers. In its canonical role, p16 inhibits the G1-S phase cell cycle progression through suppression of cyclin dependent kinases. Interestingly, p16 also has roles in metabolic reprogramming, and we previously published that loss of p16 promotes nucleotide synthesis via the pentose phosphate pathway. Whether other nucleotide metabolic genes and pathways are affected by p16/CDKN2A loss and if these can be specifically targeted in p16/CDKN2A-low tumors has not been previously explored. Using CRISPR KO libraries in multiple isogenic human and mouse melanoma cell lines, we determined that many nucleotide metabolism genes are negatively enriched in p16/CDKN2A knockdown cells compared to controls. Indeed, many of the genes that are required for survival in the context of low p16/CDKN2A expression based on our CRISPR screens are upregulated in p16 knockdown melanoma cells and those with endogenously low CDKN2A expression. We determined that cells with low p16/Cdkn2a expression are sensitive to multiple inhibitors of de novo purine synthesis, including anti-folates. Tumors with p16 knockdown were more sensitive to the anti-folate methotrexate in vivo than control tumors. Together, our data provide evidence to reevaluate the utility of these drugs in patients with p16/CDKN2A-low tumors as loss of p16/CDKN2A may provide a therapeutic window for these agents.
Collapse
Affiliation(s)
- Naveen Kumar Tangudu
- Department of Pharmacology & Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Raquel Buj
- Department of Pharmacology & Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Hui Wang
- Department of Pharmacology & Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Jiefei Wang
- Department of Biomedical Informatics and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Aidan R. Cole
- Department of Pharmacology & Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Apoorva Uboveja
- Department of Pharmacology & Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Richard Fang
- Department of Pharmacology & Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Amandine Amalric
- Department of Pharmacology & Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Peter Sajjakulnukit
- Department of Molecular and Integrative Physiology, Department of Internal Medicine, Division of Gastroenterology, and Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Maureen A. Lyons
- Genomics Facility UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Kristine Cooper
- Biostatistics Facility UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Nadine Hempel
- Division of Hematology/Oncology, Department of Medicine, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA
| | - Nathaniel W. Snyder
- Department of Cardiovascular Sciences, Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Costas A. Lyssiotis
- Department of Molecular and Integrative Physiology, Department of Internal Medicine, Division of Gastroenterology, and Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Uma R. Chandran
- Department of Biomedical Informatics and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Katherine M. Aird
- Department of Pharmacology & Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
410
|
Mathias C, Kozak VN, Magno JM, Baal SCS, dos Santos VHA, Ribeiro EMDSF, Gradia DF, Castro MAA, Carvalho de Oliveira J. PD-1/PD-L1 Inhibitors Response in Triple-Negative Breast Cancer: Can Long Noncoding RNAs Be Associated? Cancers (Basel) 2023; 15:4682. [PMID: 37835376 PMCID: PMC10572024 DOI: 10.3390/cancers15194682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 08/25/2023] [Accepted: 08/25/2023] [Indexed: 10/15/2023] Open
Abstract
As immune checkpoint inhibitors (ICI) emerge as a paradigm-shifting treatment option for patients with advanced or metastatic cancer, there is a growing demand for biomarkers that can distinguish which patients are likely to benefit. In the case of triple-negative breast cancer (TNBC), characterized by a lack of therapeutic targets, pembrolizumab approval for high-risk early-stage disease occurred regardless of PD-L1 status, which keeps the condition in a biomarker limbus. In this review, we highlight the participation of long non-coding RNAs (lncRNAs) in the regulation of the PD-1/PD-L1 pathway, as well as in the definition of prognostic immune-related signatures in many types of tumors, aiming to shed light on molecules that deserve further investigation for a potential role as biomarkers. We also conducted a bioinformatic analysis to investigate lncRNAs already investigated in PD-1/PDL-1 pathways in other cancer types, considering the TNBC molecular context. In this sense, from the generated data, we evidence here two lncRNAs, UCA1 and HCP5, which have not yet been identified in the context of the tumoral immune response in breast cancer. These candidates can be further explored to verify their use as biomarkers for ICI response. In this article, we present an updated review regarding the use of lncRNA as biomarkers of response to ICI, highlighting the versatility of using these molecules.
Collapse
Affiliation(s)
- Carolina Mathias
- Post-Graduation Program in Genetics, Department of Genetics, Federal University of Parana, Curitiba 81530-980, Brazil; (C.M.)
| | - Vanessa Nascimento Kozak
- Post-Graduation Program in Genetics, Department of Genetics, Federal University of Parana, Curitiba 81530-980, Brazil; (C.M.)
| | - Jessica Maria Magno
- Post-Graduation Program in Bioinformatics, Bioinformatics and Systems Biology Laboratory, Federal University of Paraná, Curitiba 81520-260, Brazil (V.H.A.d.S.)
| | - Suelen Cristina Soares Baal
- Post-Graduation Program in Genetics, Department of Genetics, Federal University of Parana, Curitiba 81530-980, Brazil; (C.M.)
| | - Victor Henrique Apolonio dos Santos
- Post-Graduation Program in Bioinformatics, Bioinformatics and Systems Biology Laboratory, Federal University of Paraná, Curitiba 81520-260, Brazil (V.H.A.d.S.)
| | | | - Daniela Fiori Gradia
- Post-Graduation Program in Genetics, Department of Genetics, Federal University of Parana, Curitiba 81530-980, Brazil; (C.M.)
| | - Mauro Antonio Alves Castro
- Post-Graduation Program in Bioinformatics, Bioinformatics and Systems Biology Laboratory, Federal University of Paraná, Curitiba 81520-260, Brazil (V.H.A.d.S.)
| | - Jaqueline Carvalho de Oliveira
- Post-Graduation Program in Genetics, Department of Genetics, Federal University of Parana, Curitiba 81530-980, Brazil; (C.M.)
| |
Collapse
|
411
|
Horiguchi H, Kadomatsu T, Yamashita T, Yumoto S, Terada K, Sato M, Morinaga J, Miyata K, Oike Y. ANGPTL2 promotes immune checkpoint inhibitor-related murine autoimmune myocarditis. Commun Biol 2023; 6:965. [PMID: 37736764 PMCID: PMC10517162 DOI: 10.1038/s42003-023-05338-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 09/08/2023] [Indexed: 09/23/2023] Open
Abstract
Use of immune checkpoint inhibitors (ICIs) as cancer immunotherapy advances rapidly in the clinic. Despite their therapeutic benefits, ICIs can cause clinically significant immune-related adverse events (irAEs), including myocarditis. However, the cellular and molecular mechanisms regulating irAE remain unclear. Here, we investigate the function of Angiopoietin-like protein 2 (ANGPTL2), a potential inflammatory mediator, in a mouse model of ICI-related autoimmune myocarditis. ANGPTL2 deficiency attenuates autoimmune inflammation in these mice, an outcome associated with decreased numbers of T cells and macrophages. We also show that cardiac fibroblasts express abundant ANGPTL2. Importantly, cardiac myofibroblast-derived ANGPTL2 enhances expression of chemoattractants via the NF-κB pathway, accelerating T cell recruitment into heart tissues. Our findings suggest an immunostimulatory function for ANGPTL2 in the context of ICI-related autoimmune inflammation and highlight the pathophysiological significance of ANGPTL2-mediated cardiac myofibroblast/immune cell crosstalk in enhancing autoimmune responses. These findings overall provide insight into mechanisms regulating irAEs.
Collapse
Affiliation(s)
- Haruki Horiguchi
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, 860-8556, Japan
- Department of Aging and Geriatric Medicine, Graduate School of Medical Science, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Tsuyoshi Kadomatsu
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, 860-8556, Japan.
- Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto, 860-8556, Japan.
| | - Tomoya Yamashita
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Shinsei Yumoto
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, 860-8556, Japan
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Kazutoyo Terada
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, 860-8556, Japan
- Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Michio Sato
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Jun Morinaga
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Keishi Miyata
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, 860-8556, Japan
- Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Yuichi Oike
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, 860-8556, Japan.
- Department of Aging and Geriatric Medicine, Graduate School of Medical Science, Kumamoto University, Kumamoto, 860-8556, Japan.
- Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto, 860-8556, Japan.
| |
Collapse
|
412
|
Rausch J, Ullrich E, Kühn MW. Epigenetic targeting to enhance acute myeloid leukemia-directed immunotherapy. Front Immunol 2023; 14:1269012. [PMID: 37809078 PMCID: PMC10556528 DOI: 10.3389/fimmu.2023.1269012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 09/05/2023] [Indexed: 10/10/2023] Open
Abstract
AML is a malignant disease of hematopoietic progenitor cells with unsatisfactory treatment outcome, especially in patients that are ineligible for intensive chemotherapy. Immunotherapy, comprising checkpoint inhibition, T-cell engaging antibody constructs, and cellular therapies, has dramatically improved the outcome of patients with solid tumors and lymphatic neoplasms. In AML, these approaches have been far less successful. Discussed reasons are the relatively low mutational burden of AML blasts and the difficulty in defining AML-specific antigens not expressed on hematopoietic progenitor cells. On the other hand, epigenetic dysregulation is an essential driver of leukemogenesis, and non-selective hypomethylating agents (HMAs) are the current backbone of non-intensive treatment. The first clinical trials that evaluated whether HMAs may improve immune checkpoint inhibitors' efficacy showed modest efficacy except for the anti-CD47 antibody that was substantially more efficient against AML when combined with azacitidine. Combining bispecific antibodies or cellular treatments with HMAs is subject to ongoing clinical investigation, and efficacy data are awaited shortly. More selective second-generation inhibitors targeting specific chromatin regulators have demonstrated promising preclinical activity against AML and are currently evaluated in clinical trials. These drugs that commonly cause leukemia cell differentiation potentially sensitize AML to immune-based treatments by co-regulating immune checkpoints, providing a pro-inflammatory environment, and inducing (neo)-antigen expression. Combining selective targeted epigenetic drugs with (cellular) immunotherapy is, therefore, a promising approach to avoid unintended effects and augment efficacy. Future studies will provide detailed information on how these compounds influence specific immune functions that may enable translation into clinical assessment.
Collapse
Affiliation(s)
- Johanna Rausch
- Department of Hematology and Medical Oncology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
- German Cancer Consortium (DKTK) Partner Site Frankfurt/Mainz and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Evelyn Ullrich
- German Cancer Consortium (DKTK) Partner Site Frankfurt/Mainz and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Children’s Hospital, Experimental Immunology, Johann Wolfgang Goethe University, Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe University, Frankfurt, Germany
- University Cancer Center (UCT), Frankfurt, Germany
| | - Michael W.M. Kühn
- Department of Hematology and Medical Oncology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
- German Cancer Consortium (DKTK) Partner Site Frankfurt/Mainz and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
413
|
Saad ED, Coart E, Deltuvaite-Thomas V, Garcia-Barrado L, Burzykowski T, Buyse M. Trial Design for Cancer Immunotherapy: A Methodological Toolkit. Cancers (Basel) 2023; 15:4669. [PMID: 37760636 PMCID: PMC10527464 DOI: 10.3390/cancers15184669] [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: 06/12/2023] [Revised: 08/12/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
Immunotherapy with checkpoint inhibitors (CPIs) and cell-based products has revolutionized the treatment of various solid tumors and hematologic malignancies. These agents have shown unprecedented response rates and long-term benefits in various settings. These clinical advances have also pointed to the need for new or adapted approaches to trial design and assessment of efficacy and safety, both in the early and late phases of drug development. Some of the conventional statistical methods and endpoints used in other areas of oncology appear to be less appropriate in immuno-oncology. Conversely, other methods and endpoints have emerged as alternatives. In this article, we discuss issues related to trial design in the early and late phases of drug development in immuno-oncology, with a focus on CPIs. For early trials, we review the most salient issues related to dose escalation, use and limitations of tumor response and progression criteria for immunotherapy, the role of duration of response as an endpoint in and of itself, and the need to conduct randomized trials as early as possible in the development of new therapies. For late phases, we discuss the choice of primary endpoints for randomized trials, review the current status of surrogate endpoints, and discuss specific statistical issues related to immunotherapy, including non-proportional hazards in the assessment of time-to-event endpoints, alternatives to the Cox model in these settings, and the method of generalized pairwise comparisons, which can provide a patient-centric assessment of clinical benefit and be used to design randomized trials.
Collapse
Affiliation(s)
- Everardo D. Saad
- International Drug Development Institute, Louvain-la-Neuve (IDDI), 1340 Ottignies-Louvain-la-Neuve, Belgium; (E.C.); (V.D.-T.); (L.G.-B.); (T.B.); (M.B.)
| | - Elisabeth Coart
- International Drug Development Institute, Louvain-la-Neuve (IDDI), 1340 Ottignies-Louvain-la-Neuve, Belgium; (E.C.); (V.D.-T.); (L.G.-B.); (T.B.); (M.B.)
| | - Vaiva Deltuvaite-Thomas
- International Drug Development Institute, Louvain-la-Neuve (IDDI), 1340 Ottignies-Louvain-la-Neuve, Belgium; (E.C.); (V.D.-T.); (L.G.-B.); (T.B.); (M.B.)
| | - Leandro Garcia-Barrado
- International Drug Development Institute, Louvain-la-Neuve (IDDI), 1340 Ottignies-Louvain-la-Neuve, Belgium; (E.C.); (V.D.-T.); (L.G.-B.); (T.B.); (M.B.)
| | - Tomasz Burzykowski
- International Drug Development Institute, Louvain-la-Neuve (IDDI), 1340 Ottignies-Louvain-la-Neuve, Belgium; (E.C.); (V.D.-T.); (L.G.-B.); (T.B.); (M.B.)
- Interuniversity Institute for Biostatistics and Statistical Bioinformatics (I-BioStat), Hasselt University, B-3500 Hasselt, Belgium
| | - Marc Buyse
- International Drug Development Institute, Louvain-la-Neuve (IDDI), 1340 Ottignies-Louvain-la-Neuve, Belgium; (E.C.); (V.D.-T.); (L.G.-B.); (T.B.); (M.B.)
- Interuniversity Institute for Biostatistics and Statistical Bioinformatics (I-BioStat), Hasselt University, B-3500 Hasselt, Belgium
| |
Collapse
|
414
|
Zhou X, Iwama S, Kobayashi T, Ando M, Arima H. Risk of Thyroid Dysfunction in PD-1 Blockade Is Stratified by the Pattern of TgAb and TPOAb Positivity at Baseline. J Clin Endocrinol Metab 2023; 108:e1056-e1062. [PMID: 37084392 DOI: 10.1210/clinem/dgad231] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 04/01/2023] [Accepted: 04/17/2023] [Indexed: 04/23/2023]
Abstract
CONTEXT Positive antithyroglobulin (TgAb) and/or antithyroid peroxidase antibodies (TPOAb) at baseline indicate a high risk of thyroid immune-related adverse events (irAEs) induced by antiprogrammed cell death-1 antibodies (anti-PD-1-Ab). However, whether the positivity patterns of both antibodies are associated with the risk of thyroid irAEs is unknown. OBJECTIVE The aim of the present study was to clarify the association of the pattern of TgAb and TPOAb positivity at baseline with the risk of thyroid irAEs induced by anti-PD-1-Ab. METHODS Patients (n = 516) were evaluated for TgAb and TPOAb at baseline and prospectively for thyroid function every 6 weeks for 24 weeks after initiating anti-PD-1-Ab. RESULTS Fifty-one (9.9%) patients developed thyroid irAEs (thyrotoxicosis in 34, hypothyroidism without prior thyrotoxicosis in 17). Twenty-five patients subsequently developed hypothyroidism following thyrotoxicosis. The cumulative incidence of thyroid irAEs differed among 4 groups classified by the presence of TgAb/TPOAb at baseline (group 1: TgAb-(-)/TPOAb-(-), 4.6% [19/415]; group 2: TgAb-(-)/TPOAb-(+), 15.8% [9/57]; group 3: TgAb-(+)/TPOAb-(-), 42.1% [8/19]; group 4: TgAb-(+)/TPOAb-(+), 60.0% [15/25]) as follows: groups 1 vs 2-4 (P ≤ .001) and groups 2 vs 3 (P = .008) and 4 (P < .001). There were different incidences of thyrotoxicosis (groups 1-4, 3.1%, 5.3%, 31.6%, 48.0%, respectively; P < .001) in groups 1 vs 3 and 4, and groups 2 vs 3 and 4, and of hypothyroidism (groups 1-4: 2.9%, 15.8%, 31.6%, 60.0%, respectively; P < .001) in groups 1 vs 2 to 4, and groups 2 vs 4. CONCLUSION The risk of thyroid irAEs was affected by the pattern of TgAb and TPOAb positivity at baseline; there were high risks of thyrotoxicosis in patients with TgAb-(+) and of hypothyroidism in patients with TgAb-(+) and those with TPOAb-(+).
Collapse
Affiliation(s)
- Xin Zhou
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya 466-8550, Japan
| | - Shintaro Iwama
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya 466-8550, Japan
| | - Tomoko Kobayashi
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya 466-8550, Japan
| | - Masahiko Ando
- Center for Advanced Medicine and Clinical Research, Nagoya University Hospital, Showa-ku, Nagoya 466-8550, Japan
| | - Hiroshi Arima
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya 466-8550, Japan
| |
Collapse
|
415
|
Borys SM, Reilly SP, Magill I, Zemmour D, Brossay L. PD-1 Mediated Regulation of Unique Activated CD8 + T Cells by NK Cells in the Submandibular Gland. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.15.557930. [PMID: 37745414 PMCID: PMC10515922 DOI: 10.1101/2023.09.15.557930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
The increasing utilization of anti-PD-1 immune checkpoint blockade (ICB) has led to the emergence of immune-related adverse events (irAEs), including sicca syndrome. Interestingly, we found that the submandibular gland (SMG) of PD-1 deficient mice harbors a large population of CD8 + T cells, reminiscing ICB induced sicca. This phenotype was also observed in the SMG of both NK cell-depleted C57BL/6 animals and NK cell-deficient animals. Mechanistically, using mice conditionally deficient for PD-L1 in the NK cell lineage, we discovered that NK cells regulate CD8 + T cell homeostasis via the PD-1/PD-L1 axis in this organ. Importantly, single-cell RNA sequencing of PD-1 deficient SMG CD8 + T cells reveals a unique transcriptional profile consistent with TCR activation. These cells have limited TCR diversity and phenotypically overlap with GzmK + CD8 + T autoimmune cells identified in primary Sjögren's syndrome patients. These insights into NK cell immunoregulation in the SMG, and the consequences of disrupted CD8 + T cell homeostasis, provide opportunities for preventing the development of irAEs. Highlights Elevated CD8 + T cells in the submandibular gland (SMG) of PD-1 deficient mice parallel sicca-like irAEs seen in ICB patients. In addition to their previously described hyporesponsive phenotype, NK cells in the SMG regulate CD8 + T cell homeostasis through the PD-L1/PD-1 axis. PD-1 deficient SMG CD8 + T cells display unique transcriptional profiles associated with proinflammatory functions, TCR activation, interferon stimulation, and exhaustion. Oligoclonal expansion and similarities in TCR sequences indicate T cell activation and a preference for recognizing specific antigens.
Collapse
|
416
|
Maeoka R, Nakazawa T, Matsuda R, Morimoto T, Shida Y, Yamada S, Nishimura F, Nakamura M, Nakagawa I, Park YS, Tsujimura T, Nakase H. Therapeutic Anti-KIR Antibody of 1-7F9 Attenuates the Antitumor Effects of Expanded and Activated Human Primary Natural Killer Cells on In Vitro Glioblastoma-like Cells and Orthotopic Tumors Derived Therefrom. Int J Mol Sci 2023; 24:14183. [PMID: 37762486 PMCID: PMC10531877 DOI: 10.3390/ijms241814183] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Glioblastoma (GBM) is the leading malignant intracranial tumor, where prognosis for which has remained extremely poor for two decades. Immunotherapy has recently drawn attention as a cancer treatment, including for GBM. Natural killer (NK) cells are immune cells that attack cancer cells directly and produce antitumor immunity-related cytokines. The adoptive transfer of expanded and activated NK cells is expected to be a promising GBM immunotherapy. We previously established an efficient expansion method that produced highly purified, activated primary human NK cells, which we designated genuine induced NK cells (GiNKs). The GiNKs demonstrated antitumor effects in vitro and in vivo, which were less affected by blockade of the inhibitory checkpoint receptor programmed death 1 (PD-1). In the present study, we assessed the antitumor effects of GiNKs, both alone and combined with an antibody targeting killer Ig-like receptor 2DLs (KIR2DL1 and DL2/3, both inhibitory checkpoint receptors of NK cells) in vitro and in vivo with U87MG GBM-like cells and the T98G GBM cell line. Impedance-based real-time cell growth assays and apoptosis detection assays revealed that the GiNKs exhibited growth inhibitory effects on U87MG and T98G cells by inducing apoptosis. KIR2DL1 blockade attenuated the growth inhibition of the cell lines in vitro. The intracranial administration of GiNKs prolonged the overall survival of the U87MG-derived orthotopic xenograft brain tumor model. The KIR2DL1 blockade did not enhance the antitumor effects; rather, it attenuated it in the same manner as in the in vitro experiment. GiNK immunotherapy directly administered to the brain could be a promising immunotherapeutic alternative for patients with GBM. Furthermore, KIR2DL1 blockade appeared to require caution when used concomitantly with GiNKs.
Collapse
Affiliation(s)
- Ryosuke Maeoka
- Department of Neurosurgery, Nara Medical University, Nara 634-8521, Japan; (R.M.); (T.M.); (Y.S.); (S.Y.); (F.N.); (M.N.); (I.N.); (Y.-S.P.); (H.N.)
| | - Tsutomu Nakazawa
- Department of Neurosurgery, Nara Medical University, Nara 634-8521, Japan; (R.M.); (T.M.); (Y.S.); (S.Y.); (F.N.); (M.N.); (I.N.); (Y.-S.P.); (H.N.)
- Grandsoul Research Institute for Immunology, Inc., Uda 633-2221, Japan;
- Clinic Grandsoul Nara, Uda 633-2221, Japan
| | - Ryosuke Matsuda
- Department of Neurosurgery, Nara Medical University, Nara 634-8521, Japan; (R.M.); (T.M.); (Y.S.); (S.Y.); (F.N.); (M.N.); (I.N.); (Y.-S.P.); (H.N.)
| | - Takayuki Morimoto
- Department of Neurosurgery, Nara Medical University, Nara 634-8521, Japan; (R.M.); (T.M.); (Y.S.); (S.Y.); (F.N.); (M.N.); (I.N.); (Y.-S.P.); (H.N.)
| | - Yoichi Shida
- Department of Neurosurgery, Nara Medical University, Nara 634-8521, Japan; (R.M.); (T.M.); (Y.S.); (S.Y.); (F.N.); (M.N.); (I.N.); (Y.-S.P.); (H.N.)
| | - Shuichi Yamada
- Department of Neurosurgery, Nara Medical University, Nara 634-8521, Japan; (R.M.); (T.M.); (Y.S.); (S.Y.); (F.N.); (M.N.); (I.N.); (Y.-S.P.); (H.N.)
| | - Fumihiko Nishimura
- Department of Neurosurgery, Nara Medical University, Nara 634-8521, Japan; (R.M.); (T.M.); (Y.S.); (S.Y.); (F.N.); (M.N.); (I.N.); (Y.-S.P.); (H.N.)
| | - Mitsutoshi Nakamura
- Department of Neurosurgery, Nara Medical University, Nara 634-8521, Japan; (R.M.); (T.M.); (Y.S.); (S.Y.); (F.N.); (M.N.); (I.N.); (Y.-S.P.); (H.N.)
- Clinic Grandsoul Nara, Uda 633-2221, Japan
| | - Ichiro Nakagawa
- Department of Neurosurgery, Nara Medical University, Nara 634-8521, Japan; (R.M.); (T.M.); (Y.S.); (S.Y.); (F.N.); (M.N.); (I.N.); (Y.-S.P.); (H.N.)
| | - Young-Soo Park
- Department of Neurosurgery, Nara Medical University, Nara 634-8521, Japan; (R.M.); (T.M.); (Y.S.); (S.Y.); (F.N.); (M.N.); (I.N.); (Y.-S.P.); (H.N.)
| | - Takahiro Tsujimura
- Grandsoul Research Institute for Immunology, Inc., Uda 633-2221, Japan;
- Clinic Grandsoul Nara, Uda 633-2221, Japan
| | - Hiroyuki Nakase
- Department of Neurosurgery, Nara Medical University, Nara 634-8521, Japan; (R.M.); (T.M.); (Y.S.); (S.Y.); (F.N.); (M.N.); (I.N.); (Y.-S.P.); (H.N.)
| |
Collapse
|
417
|
Motoo I, Ando T, Hamashima T, Kajiura S, Sakumura M, Ueda Y, Murayama A, Ogawa K, Tsukada K, Ueda A, Suzuki N, Nakada N, Nakashima K, Hosokawa A, Yasuda I. Liver metastasis affects progression pattern during immune checkpoint inhibitors monotherapy in gastric cancer. Front Oncol 2023; 13:1193533. [PMID: 37790758 PMCID: PMC10542891 DOI: 10.3389/fonc.2023.1193533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 08/29/2023] [Indexed: 10/05/2023] Open
Abstract
Introduction The efficacy of immune checkpoint inhibitors (ICIs) is heterogeneous at each metastatic site, and tumor progression pattern is associated with survival; however, it remains unclear in gastric cancer (GC). Therefore, we aimed to clarify the progression pattern in response to ICIs in patients with GC, and we analyzed its mechanism focusing on the intratumoral immune cells. Methods Patients who received ICIs were retrospectively classified into non-systemic and systemic progression groups based on their radiological assessments. Moreover, the best percentage change in target lesions from each organ was compared. Results Among 148 patients, the non-systemic progression group showed a significant improvement in overall survival (OS) compared with the systemic progression group (median, 5.6 months vs. 3.3 months; HR, 0.53; 95%CI, 0.32-0.89; p = 0.012). Poor performance status (HR, 1.73, 95%CI, 1.00-2.87) and systemic progression (HR, 3.09, 95%CI, 1.95-4.82) were associated with OS. Of all metastatic sites, the liver showed the poorest percentage change, and liver metastasis (OR, 2.99, 95%CI, 1.04-8.58) was associated with systemic progression. Hence, intratumoral CD8+ T-cell density was lower in patients with liver metastasis than in those without liver metastasis after ICIs, although the density of CD4+ T-cells (Th1, Th17, and Treg) and CD163+ cells (TAM) were not significantly different. Conclusion The new progression pattern was associated with OS in GC. Liver metastasis may be a predictive factor of systemic progression during ICIs by regulating intratumoral CD8+ T-cells.
Collapse
Affiliation(s)
- Iori Motoo
- Third Department of Internal Medicine, University of Toyama, Toyama, Japan
| | - Takayuki Ando
- Third Department of Internal Medicine, University of Toyama, Toyama, Japan
| | | | - Shinya Kajiura
- Third Department of Internal Medicine, University of Toyama, Toyama, Japan
| | - Miho Sakumura
- Third Department of Internal Medicine, University of Toyama, Toyama, Japan
| | - Yuko Ueda
- Third Department of Internal Medicine, University of Toyama, Toyama, Japan
| | - Aiko Murayama
- Third Department of Internal Medicine, University of Toyama, Toyama, Japan
| | - Kohei Ogawa
- Department of Gastroenterology, Toyama Prefectural Central Hospital, Toyama, Japan
| | - Kenichiro Tsukada
- Department of Gastroenterology, Kouseiren Takaoka Hospital, Takaoka, Japan
| | - Akira Ueda
- Department of Medical Oncology, Toyama Red Cross Hospital, Toyama, Japan
| | - Nobuhiro Suzuki
- Department of Gastroenterology, Jouetsu Sogo Hospital, Jouetsu, Japan
| | - Naokatsu Nakada
- Department of Gastroenterology, Itoigawa Sogo Hospital, Itoigawa, Japan
| | - Koji Nakashima
- Department of Clinical Oncology, University of Miyazaki Hospital, Miyazaki, Japan
| | - Ayumu Hosokawa
- Third Department of Internal Medicine, University of Toyama, Toyama, Japan
- Department of Clinical Oncology, University of Miyazaki Hospital, Miyazaki, Japan
| | - Ichiro Yasuda
- Third Department of Internal Medicine, University of Toyama, Toyama, Japan
| |
Collapse
|
418
|
Helgadottir H, Ny L, Ullenhag GJ, Falkenius J, Mikiver R, Olofsson Bagge R, Isaksson K. Survival after introduction of adjuvant treatment in stage III melanoma: a nationwide registry-based study. J Natl Cancer Inst 2023; 115:1077-1084. [PMID: 37227040 PMCID: PMC10483326 DOI: 10.1093/jnci/djad081] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 04/25/2023] [Accepted: 05/10/2023] [Indexed: 05/26/2023] Open
Abstract
BACKGROUND Adjuvant treatments with PD-1 and BRAF+MEK inhibitors statistically significantly prolong recurrence-free survival in stage III cutaneous melanoma. Yet, the effect on overall survival is still unclear. Based on recurrence-free survival outcomes, these treatments have been approved and widely implemented. The treatments have considerable side effects and costs, and overall survival effect remains a highly anticipated outcome. METHODS Clinical and histopathological parameters were obtained from the Swedish Melanoma Registry for patients diagnosed with stage III melanoma between 2016 and 2020. The patients were divided depending on if they were diagnosed before or from July 2018, based on the timepoint when adjuvant treatment was introduced in Sweden. Patients were followed up until the end of 2021. In this cohort study, melanoma-specific and overall survival were calculated using the Kaplan-Meier method and Cox-regression analyses. RESULTS There were 1371 patients diagnosed with stage III primary melanoma in Sweden in 2016-2020. The 2-year overall survival rates, comparing the 634 patients in the precohort and the 737 in the postcohort, were 84.3% (95% confidence interval [CI] = 81.4% to 87.3%) and 86.1% (95% CI = 83.4% to 89.0%), respectively, with an adjusted hazard ratio of 0.91 (95% CI = 0.70 to 1.19, P = .51). Further, no statistically significant overall or melanoma-specific survival differences were seen when comparing the precohort and the postcohort in different subgroups for age, sex, or tumor characteristics. CONCLUSIONS In this nationwide population-based and registry-based study, no survival benefit was detected in patients diagnosed before or after the implementation of adjuvant treatment in stage III melanoma. These findings encourage a careful assessment of the current recommendations on adjuvant treatment.
Collapse
Affiliation(s)
- Hildur Helgadottir
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
- Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Lars Ny
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Gustav J Ullenhag
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Department of Oncology, Uppsala University Hospital, Uppsala, Sweden
| | - Johan Falkenius
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
- Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Rasmus Mikiver
- Regional Cancer Center Southeast Sweden and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Roger Olofsson Bagge
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Karolin Isaksson
- Department of Clinical Sciences, Surgery, Lund University, Lund, Sweden
- Department of Surgery, Kristianstad Hopsital, Kristianstad, Sweden
| |
Collapse
|
419
|
Xu C, Xie XL, Kang N, Jiang HQ. Evaluation of ITGB1 expression as a predictor of the therapeutic effects of immune checkpoint inhibitors in gastric cancer. BMC Gastroenterol 2023; 23:298. [PMID: 37667169 PMCID: PMC10478479 DOI: 10.1186/s12876-023-02930-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 08/22/2023] [Indexed: 09/06/2023] Open
Abstract
BACKGROUND Gastric cancer (CC) is a disease with high incidence and mortality rate. Immunotherapy is an important method for gastric cancer while lack of effective predictor. Integrins play an important role in the development. We aimed to explore the predictive value of β1 integrin (ITGB1) as a predictor of immunnotherapy in gastric cancer. METHODS Differential expression analysis was conducted using the Gene Expression Profiling Interactive Analysis (GEPIA) 2.0 and GEO databases. GEPIA data were used to evaluate the prognostic value of ITGB1 in gastric cancer (GC). Transcriptomic and clinical data of GC and normal tissues were downloaded from The Cancer Genome Atlas database, and the TIMER database was used to evaluate the association between ITGB1 and immune infiltration. Time-dependent receiver operating characteristic (ROC) curve analysis was used to determine the prognostic value of ITGB1. To verify ITGB1 expression at the protein level, immunohistochemical staining was conducted. In addition, to analyze the correlation of ITGB1 with PD-1 and PD-L1, we examined levels of PD-1 and PD-L1 by IHC and determined the predictive value of ITGB1 for anti-PD-1 therapy in GC by ROC curve analysis. RESULTS Compared with normal tissues, analysis of GEPIA and data at protein levels showed significantly higher expression of ITGB1 in GC. In addition, higher expression of ITGB1 was associated with worse pathological G-staging and tumor T-staging, which suggested that ITGB1 is a risk factor for poor prognosis in GC. The level of ITGB1 expression was positively correlated with CD8 + T cells, neutrophils, macrophages, and dendritic cells. ITGB1 expression was also correlated with PD-L1 expression, and this was further verified at the protein level by immunohistochemical analysis. The area under the ROC curve was 0.808. CONCLUSION ITGB1 may be a promising prognostic biomarker and effective predictor for anti-PD-1 therapy in GC. TRIAL REGISTRATION Retrospectively registered.
Collapse
Affiliation(s)
- Chao Xu
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, No. 215, Heping West Road, Shijiazhuang, 050000, Hebei Province, China
- Handan Central Hospital, No.15, Zhonghua Road, Handan, 056001, Hebei Province, China
| | - Xiao-Li Xie
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, No. 215, Heping West Road, Shijiazhuang, 050000, Hebei Province, China
| | - Ning Kang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, No. 215, Heping West Road, Shijiazhuang, 050000, Hebei Province, China
| | - Hui-Qing Jiang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, No. 215, Heping West Road, Shijiazhuang, 050000, Hebei Province, China.
| |
Collapse
|
420
|
Zhang Z, Pan Q, Lu M, Zhao B. Intermediate endpoints as surrogates for outcomes in cancer immunotherapy: a systematic review and meta-analysis of phase 3 trials. EClinicalMedicine 2023; 63:102156. [PMID: 37600482 PMCID: PMC10432823 DOI: 10.1016/j.eclinm.2023.102156] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 07/24/2023] [Accepted: 07/26/2023] [Indexed: 08/22/2023] Open
Abstract
Background Cancer immunotherapy shows unique efficacy kinetics that differs from conventional treatment. These characteristics may lead to the prolongation of trial duration, hence reliable surrogate endpoints are urgently needed. We aimed to systematically evaluate the study-level performance of commonly reported intermediate clinical endpoints for surrogacy in cancer immunotherapy. Methods We searched the Embase, PubMed, and Cochrane databases, between database inception and October 18, 2022, for phase 3 randomised trials investigating the efficacy of immunotherapy in patients with advanced solid tumours. An updated search was done on July, 15, 2023. No language restrictions were used. Eligible trials had to set overall survival (OS) as the primary or co-primary endpoint and report at least one intermediate clinical endpoint including objective response rate (ORR), disease control rate (DCR), progression-free survival (PFS), and 1-year overall survival. Other key inclusion and exclusion criteria included: (1) adult patients (>18 years old) with advanced solid tumour; (2) no immunotherapy conducted in the control arms; (3) follow-up is long enough to achieve OS; (4) data should be public available. A two-stage meta-analytic approach was conducted to evaluate the magnitude of the association between these intermediate endpoints and OS. A surrogate was identified if the coefficient of determination (R2) was 0.7 or greater. Leave-one-out cross-validation and pre-defined subgroup analysis were conducted to examine the heterogeneity. Potential publication bias was evaluated using the Egger's and Begg's tests. This trial was registered with PROSPERO, number CRD42022381648. Findings 52,342 patients with 15 types of tumours from 77 phase 3 studies were included. ORR (R2 = 0.11; 95% CI, 0.00-0.24), DCR (R2 = 0.01; 95% CI, 0.00-0.01), and PFS (R2 = 0.40; 95% CI, 0.23-0.56) showed weak associations with OS. However, a strong correlation was observed between 1-year survival and clinical outcome (R2 = 0.74; 95% CI, 0.64-0.83). These associations remained relatively consistent across pre-defined subgroups stratified based on tumour types, masking methods, line of treatments, drug targets, treatment strategies, and follow-up durations. No significant heterogeneities or publication bias were identified. Interpretation 1-year milestone survival was the only identified surrogacy endpoint for outcomes in cancer immunotherapy. Ongoing investigations and development of new endpoints and incorporation of biomarkers are needed to identify potential surrogate markers that can be more robust than 1-year survival. This work may provide important references in assisting the design and interpretation of future clinical trials, and constitute complementary information in drafting clinical practice guidelines. Funding None.
Collapse
Affiliation(s)
- Zhishan Zhang
- Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China
| | - Qunxiong Pan
- Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China
| | - Mingdong Lu
- The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Bin Zhao
- Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China
| |
Collapse
|
421
|
Pich-Bavastro C, Yerly L, Di Domizio J, Tissot-Renaud S, Gilliet M, Kuonen F. Activin A-Mediated Polarization of Cancer-Associated Fibroblasts and Macrophages Confers Resistance to Checkpoint Immunotherapy in Skin Cancer. Clin Cancer Res 2023; 29:3498-3513. [PMID: 37327314 PMCID: PMC10472111 DOI: 10.1158/1078-0432.ccr-23-0219] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 05/05/2023] [Accepted: 06/14/2023] [Indexed: 06/18/2023]
Abstract
PURPOSE Cemiplimab is approved for the treatment of locally advanced basal cell carcinomas (BCC), although with mitigated results. We sought to interrogate the cellular and molecular transcriptional reprogramming underlying BCC resistance to immunotherapy. EXPERIMENTAL DESIGN Here, we combined spatial and single-cell transcriptomics to deconvolute the spatial heterogeneity of the tumor microenvironment in regard with response to immunotherapy, in a cohort of both naïve and resistant BCCs. RESULTS We identified subsets of intermingled cancer-associated fibroblasts (CAF) and macrophages contributing the most to CD8 T-cell exclusion and immunosuppression. Within this spatially resolved peritumoral immunosuppressive niche, CAFs and adjacent macrophages were found to display Activin A-mediated transcriptional reprogramming towards extracellular matrix remodeling, suggesting active participation to CD8 T-cell exclusion. In independent datasets of human skin cancers, Activin A-conditioned CAFs and macrophages were associated with resistance to immune checkpoint inhibitors (ICI). CONCLUSIONS Altogether, our data identify the cellular and molecular plasticity of tumor microenvironment (TME) and the pivotal role of Activin A in polarizing the TME towards immune suppression and ICI resistance.
Collapse
Affiliation(s)
- Christine Pich-Bavastro
- Department of Dermatology and Venereology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Laura Yerly
- Department of Dermatology and Venereology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Jeremy Di Domizio
- Department of Dermatology and Venereology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Stéphanie Tissot-Renaud
- Department of Oncology, Immune Landscape Laboratory, Center of Experimental Therapeutics, Lausanne University Hospital, Lausanne, Switzerland
- Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Michel Gilliet
- Department of Dermatology and Venereology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - François Kuonen
- Department of Dermatology and Venereology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
422
|
Boutros C, Belkadi-Sadou D, Marchand A, Roy S, Routier E, Robert C. Cured or Not? Long-term Outcomes of Immunotherapy Responders. Focus on Melanoma. Curr Oncol Rep 2023; 25:989-996. [PMID: 37266890 DOI: 10.1007/s11912-023-01429-x] [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: 05/03/2023] [Indexed: 06/03/2023]
Abstract
PURPOSE OF REVIEW Immune checkpoint inhibitors (ICIs) demonstrated robust antitumor activity and tolerable safety in advanced melanoma. Data on long-term outcome of patients who benefited from this therapy and who are still free of progression despite ICI discontinuation is now available. We review here the characteristics of long-term ICI responders and address the critical question of cure. RECENT FINDINGS Long-term outcome of patients with metastatic melanoma enrolled in large phase 2 and phase 3 clinical trials evaluating ICI in metastatic melanoma is now available. Durable responses, with more than 6 years of median follow-up, may persist after discontinuation. They occur more frequently in patients who achieved a complete response rather than in patients who had partial response or stable disease. Although long-term clinical benefit is more frequent in patients with high PDL-1 expression and smaller tumor burden, durable response may also be observed regardless of baseline characteristics. In patients with asymptomatic brain metastasis, combined immunotherapy (ipilimumab plus nivolumab) may also lead to long-term remission. Clinical trials confirm the durable antitumor activity of ICI. Although the hope for cure seems reasonable for many patients in this situation, late relapses may occur and no relapse-predictive biomarkers have been identified yet. Long-term responders who relapse can respond to a rechallenge of ICI although data are limited concerning the rate and the duration of this new response.
Collapse
Affiliation(s)
- Céline Boutros
- Dermatology Unit, Department of Medicine, Gustave Roussy Cancer Campus, 114 Rue Edouard Vaillant, 94805, Villejuif, France
- Outpatient Clinic, Department of Medicine, Gustave Roussy Cancer Campus, Villejuif, France
| | - Djaouida Belkadi-Sadou
- Dermatology Unit, Department of Medicine, Gustave Roussy Cancer Campus, 114 Rue Edouard Vaillant, 94805, Villejuif, France
| | - Antoine Marchand
- Dermatology Unit, Department of Medicine, Gustave Roussy Cancer Campus, 114 Rue Edouard Vaillant, 94805, Villejuif, France
| | - Séverine Roy
- Dermatology Unit, Department of Medicine, Gustave Roussy Cancer Campus, 114 Rue Edouard Vaillant, 94805, Villejuif, France
| | - Emilie Routier
- Dermatology Unit, Department of Medicine, Gustave Roussy Cancer Campus, 114 Rue Edouard Vaillant, 94805, Villejuif, France
| | - Caroline Robert
- Dermatology Unit, Department of Medicine, Gustave Roussy Cancer Campus, 114 Rue Edouard Vaillant, 94805, Villejuif, France.
- University Paris-Saclay, Faculty of Medicine, Kremlin-Bicêtre, France.
- INSERM Unit U981, Villejuif, France.
| |
Collapse
|
423
|
Scobie MR, Zhou KI, Ahmed S, Kelley MJ. Utility of Tumor Mutational Burden as a Biomarker for Response to Immune Checkpoint Inhibition in the VA Population. JCO Precis Oncol 2023; 7:e2300176. [PMID: 38039430 DOI: 10.1200/po.23.00176] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 07/26/2023] [Accepted: 08/24/2023] [Indexed: 12/03/2023] Open
Abstract
PURPOSE Immune checkpoint inhibitors (ICIs) are used for an increasing number of indications across various tumor types, as well as several tumor-agnostic indications in patients with advanced cancer. Although many patients benefit from ICI therapy, others do not, highlighting a need for better predictive biomarkers. Tumor mutational burden (TMB) reflects the global number of mutations within a tumor and has been widely explored as a predictive biomarker of ICI response. The current tumor type-agnostic US Food and Drug Administration approval of pembrolizumab for metastatic solid tumors defines high TMB (TMB-H) as ≥10 mut/Mb as measured by FoundationOne CDx. This fixed cutoff may not be the ideal value across all solid tumors. METHODS We performed a retrospective analysis of the association of survival outcomes with TMB in patients treated with ICI for five major cancer types, using real-world data from the VA. Survival was measured from initiation of ICI, and Kaplan-Meier survival curves were compared by log-rank test. RESULTS Overall survival (OS) was significantly longer for patients with TMB-H versus TMB low tumors in non-small-cell lung cancer (NSCLC; n = 1,593), head and neck (H&N) cancer (n = 222), and urothelial cancer (n = 332). OS was not significantly different based on TMB status in melanoma (n = 207) or esophageal/gastric cancer (n = 248). CONCLUSION Consistent with previous studies, a predictive value of TMB ≥10 mut/Mb for ICI response was found in NSCLC and H&N, but not in esophageal/gastric cancer. Although inconclusive in the literature, significant association was found in urothelial cancer. The predictive value of TMB in melanoma was inconclusive. Our analysis does not support the use of a fixed threshold for TMB as a standalone predictive biomarker for ICI across all solid tumors.
Collapse
Affiliation(s)
- Micaela R Scobie
- Department of Veterans Affairs, National Oncology Program, Washington, DC
- Division of Hematology-Oncology, Durham VA Medical Center, Durham, NC
| | - Katherine I Zhou
- Division of Hematology-Oncology, Durham VA Medical Center, Durham, NC
- Division of Medical Oncology, Department of Medicine, Duke University, Durham, NC
| | - Sara Ahmed
- Department of Veterans Affairs, National Oncology Program, Washington, DC
| | - Michael J Kelley
- Department of Veterans Affairs, National Oncology Program, Washington, DC
- Division of Hematology-Oncology, Durham VA Medical Center, Durham, NC
| |
Collapse
|
424
|
Wang J, Lee CS, Attarian S, Kohn N, Devoe C. Inpatient utilization of immune checkpoint inhibitors and clinical outcomes. J Oncol Pharm Pract 2023; 29:1392-1397. [PMID: 36131556 DOI: 10.1177/10781552221123967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
INTRODUCTION Retrospective studies have suggested that patients with poor performance status treated with immune checkpoint inhibitors have shorter overall survival and poorer response rates. This study was undertaken to investigate the possible relationships between inpatient immune checkpoint inhibitor use and clinical outcomes. METHODS This was a retrospective chart review of cancer patients who received an immune checkpoint inhibitor while hospitalized from 1 January 2016 to 30 December 2020. The primary outcome was 90-day mortality or admission to hospice rate. Secondary outcomes included overall survival, time to death or discharge to hospice, and descriptive summarization of patient characteristics. RESULTS A total of 52 patients were analyzed. At 90 days, 68.2% of subjects were expired or admitted to hospice (95% CI: 54.7-81%). 90-day overall survival was 47.1%; median survival time was 81 days (95% CI: 28-242 days). The median time to death or hospice was 35 days (95% CI: 24-72 days). The time to death or hospice was shorter for immune checkpoint inhibitor-naive patients compared to those who received immune checkpoint inhibitors prior to admission (29 days, 95% CI: 12-43 days vs. 242 days, 95% CI: 36-1288 days, respectively; HR: 2.74, 95% CI: 1.2-6.25; p = 0.0121). No differences were found when comparing other baseline characteristics. CONCLUSION A majority of patients who received an immune checkpoint inhibitor while hospitalized were either discharged to hospice or expired by 90 days. An increased rate of death or discharge to hospice was observed for patients who were immune checkpoint inhibitor-naive prior to their admission.
Collapse
Affiliation(s)
| | | | | | - Nina Kohn
- Biostatistics Unit, Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | | |
Collapse
|
425
|
Wang X, Jiang Y, Chen H, Zhang T, Han Z, Chen C, Yuan Q, Xiong W, Wang W, Li G, Heng PA, Li R. Cancer immunotherapy response prediction from multi-modal clinical and image data using semi-supervised deep learning. Radiother Oncol 2023; 186:109793. [PMID: 37414254 DOI: 10.1016/j.radonc.2023.109793] [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: 01/20/2023] [Revised: 06/22/2023] [Accepted: 06/28/2023] [Indexed: 07/08/2023]
Abstract
BACKGROUND AND PURPOSE Immunotherapy is a standard treatment for many tumor types. However, only a small proportion of patients derive clinical benefit and reliable predictive biomarkers of immunotherapy response are lacking. Although deep learning has made substantial progress in improving cancer detection and diagnosis, there is limited success on the prediction of treatment response. Here, we aim to predict immunotherapy response of gastric cancer patients using routinely available clinical and image data. MATERIALS AND METHODS We present a multi-modal deep learning radiomics approach to predict immunotherapy response using both clinical data and computed tomography images. The model was trained using 168 advanced gastric cancer patients treated with immunotherapy. To overcome limitations of small training data, we leverage an additional dataset of 2,029 patients who did not receive immunotherapy in a semi-supervised framework to learn intrinsic imaging phenotypes of the disease. We evaluated model performance in two independent cohorts of 81 patients treated with immunotherapy. RESULTS The deep learning model achieved area under receiver operating characteristics curve (AUC) of 0.791 (95% CI 0.633-0.950) and 0.812 (95% CI 0.669-0.956) for predicting immunotherapy response in the internal and external validation cohorts. When combined with PD-L1 expression, the integrative model further improved the AUC by 4-7% in absolute terms. CONCLUSION The deep learning model achieved promising performance for predicting immunotherapy response from routine clinical and image data. The proposed multi-modal approach is general and can incorporate other relevant information to further improve prediction of immunotherapy response.
Collapse
Affiliation(s)
- Xi Wang
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford 94305, CA, USA; Department of Computer Science and Engineering, The Chinese University of Hong Kong, Hong Kong, China; Zhejiang Lab, Hangzhou, China
| | - Yuming Jiang
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford 94305, CA, USA; Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Hao Chen
- Department of Computer Science and Engineering, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Taojun Zhang
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Zhen Han
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Chuanli Chen
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qingyu Yuan
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wenjun Xiong
- Department of Gastrointestinal Surgery, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wei Wang
- Department of Gastric Surgery, and State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Guoxin Li
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Pheng-Ann Heng
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, Hong Kong, China
| | - Ruijiang Li
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford 94305, CA, USA.
| |
Collapse
|
426
|
VanderWalde A, Bellasea SL, Kendra KL, Khushalani NI, Campbell KM, Scumpia PO, Kuklinski LF, Collichio F, Sosman JA, Ikeguchi A, Victor AI, Truong TG, Chmielowski B, Portnoy DC, Chen Y, Margolin K, Bane C, Dasanu CA, Johnson DB, Eroglu Z, Chandra S, Medina E, Gonzalez CR, Baselga-Carretero I, Vega-Crespo A, Garcilazo IP, Sharon E, Hu-Lieskovan S, Patel SP, Grossmann KF, Moon J, Wu MC, Ribas A. Ipilimumab with or without nivolumab in PD-1 or PD-L1 blockade refractory metastatic melanoma: a randomized phase 2 trial. Nat Med 2023; 29:2278-2285. [PMID: 37592104 PMCID: PMC10708907 DOI: 10.1038/s41591-023-02498-y] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 07/12/2023] [Indexed: 08/19/2023]
Abstract
In this randomized phase 2 trial, blockade of cytotoxic T-lymphocyte protein 4 (CTLA-4) with continuation of programmed death protein 1 (PD-1) blockade in patients with metastatic melanoma who had received front-line anti-PD-1 or therapy against programmed cell death 1 ligand 1 and whose tumors progressed was tested in comparison with CTLA-4 blockade alone. Ninety-two eligible patients were randomly assigned in a 3:1 ratio to receive the combination of ipilimumab and nivolumab, or ipilimumab alone. The primary endpoint was progression-free survival. Secondary endpoints included the difference in CD8 T cell infiltrate among responding and nonresponding tumors, objective response rate, overall survival and toxicity. The combination of nivolumab and ipilimumab resulted in a statistically significant improvement in progression-free survival over ipilimumab (hazard ratio = 0.63, 90% confidence interval (CI) = 0.41-0.97, one-sided P = 0.04). Objective response rates were 28% (90% CI = 19-38%) and 9% (90% CI = 2-25%), respectively (one-sided P = 0.05). Grade 3 or higher treatment-related adverse events occurred in 57% and 35% of patients, respectively, which is consistent with the known toxicity profile of these regimens. The change in intratumoral CD8 T cell density observed in the present analysis did not reach statistical significance to support the formal hypothesis tested as a secondary endpoint. In conclusion, primary resistance to PD-1 blockade therapy can be reversed in some patients with the combination of CTLA-4 and PD-1 blockade. Clinicaltrials.gov identifier: NCT03033576 .
Collapse
Affiliation(s)
| | - Shay L Bellasea
- Southwest Oncology Group Statistics and Data Management Center, Seattle, WA, USA
- Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Kari L Kendra
- Ohio State University Wexner Medical Center, Columbus, OH, USA
| | | | - Katie M Campbell
- Jonsson Comprehensive Cancer Center at the University of California, Los Angeles, CA, USA
| | - Philip O Scumpia
- Jonsson Comprehensive Cancer Center at the University of California, Los Angeles, CA, USA
| | - Lawrence F Kuklinski
- Jonsson Comprehensive Cancer Center at the University of California, Los Angeles, CA, USA
| | - Frances Collichio
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
| | - Jeffrey A Sosman
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| | | | | | - Thach-Giao Truong
- Kaiser Permanente Northern California, Kaiser Permanente National Cancer Institute Community Oncology Research Program, Vallejo, CA, USA
| | - Bartosz Chmielowski
- Jonsson Comprehensive Cancer Center at the University of California, Los Angeles, CA, USA
| | | | - Yuanbin Chen
- Cancer and Hematology Centers of Western Michigan-Cancer Research Consortium of West Michigan, Grand Rapids, MI, USA
| | - Kim Margolin
- City of Hope Comprehensive Cancer Center, Duarte, CA, USA
- St. John's Cancer Institute, Santa Monica, CA, USA
| | - Charles Bane
- Dayton Physicians LLC, Miami Valley Hospital North, Dayton, OH, USA
| | | | | | | | - Sunandana Chandra
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| | - Egmidio Medina
- Jonsson Comprehensive Cancer Center at the University of California, Los Angeles, CA, USA
| | - Cynthia R Gonzalez
- Jonsson Comprehensive Cancer Center at the University of California, Los Angeles, CA, USA
| | | | - Agustin Vega-Crespo
- Jonsson Comprehensive Cancer Center at the University of California, Los Angeles, CA, USA
| | - Ivan Perez Garcilazo
- Jonsson Comprehensive Cancer Center at the University of California, Los Angeles, CA, USA
| | - Elad Sharon
- National Cancer Institute, Cancer Therapy Evaluation Program, Bethesda, MD, USA
| | | | - Sapna P Patel
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kenneth F Grossmann
- University of Utah, Huntsman Cancer Institute, Salt Lake City, UT, USA
- Merck & Co., Inc., Rahway, NJ, USA
| | - James Moon
- Southwest Oncology Group Statistics and Data Management Center, Seattle, WA, USA
- Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Michael C Wu
- Southwest Oncology Group Statistics and Data Management Center, Seattle, WA, USA
- Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Antoni Ribas
- Jonsson Comprehensive Cancer Center at the University of California, Los Angeles, CA, USA.
| |
Collapse
|
427
|
Yang K, Halima A, Chan TA. Antigen presentation in cancer - mechanisms and clinical implications for immunotherapy. Nat Rev Clin Oncol 2023; 20:604-623. [PMID: 37328642 DOI: 10.1038/s41571-023-00789-4] [Citation(s) in RCA: 97] [Impact Index Per Article: 48.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/25/2023] [Indexed: 06/18/2023]
Abstract
Over the past decade, the emergence of effective immunotherapies has revolutionized the clinical management of many types of cancers. However, long-term durable tumour control is only achieved in a fraction of patients who receive these therapies. Understanding the mechanisms underlying clinical response and resistance to treatment is therefore essential to expanding the level of clinical benefit obtained from immunotherapies. In this Review, we describe the molecular mechanisms of antigen processing and presentation in tumours and their clinical consequences. We examine how various aspects of the antigen-presentation machinery (APM) shape tumour immunity. In particular, we discuss genomic variants in HLA alleles and other APM components, highlighting their influence on the immunopeptidomes of both malignant cells and immune cells. Understanding the APM, how it is regulated and how it changes in tumour cells is crucial for determining which patients will respond to immunotherapy and why some patients develop resistance. We focus on recently discovered molecular and genomic alterations that drive the clinical outcomes of patients receiving immune-checkpoint inhibitors. An improved understanding of how these variables mediate tumour-immune interactions is expected to guide the more precise administration of immunotherapies and reveal potentially promising directions for the development of new immunotherapeutic approaches.
Collapse
Affiliation(s)
- Kailin Yang
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA
| | - Ahmed Halima
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA
| | - Timothy A Chan
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA.
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA.
- National Center for Regenerative Medicine, Cleveland, OH, USA.
- Case Comprehensive Cancer Center, Cleveland, OH, USA.
| |
Collapse
|
428
|
Berner F, Flatz L. Autoimmunity in immune checkpoint inhibitor-induced immune-related adverse events: A focus on autoimmune skin toxicity and pneumonitis. Immunol Rev 2023; 318:37-50. [PMID: 37548043 DOI: 10.1111/imr.13258] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 06/29/2023] [Indexed: 08/08/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized cancer therapy. However, their use is frequently associated with immune-related adverse events (irAEs) potentially affecting any organ. The mechanisms mediating such irAEs remain poorly understood and biomarkers to predict the development of irAEs are lacking. Growing evidence shows the importance of self-antigens in mediating irAEs during ICI therapy, in particular the well-described melanocyte differentiation antigens in melanoma patients. This review will focus on two novel classes of self-antigens involved in mediating autoimmune skin toxicity and pneumonitis in non-small cell lung cancer patients treated with immunotherapy. T cells specific for these self-antigens are thought to not only mediate irAEs but are thought to simultaneously mediate anti-tumor responses and are therefore associated with both autoimmune toxicity and response to ICI therapy. We further discuss emerging cellular and proteomic immune signatures of irAEs that may serve as biomarkers to help predict which patients are at higher risk of developing these irAEs. The determination of new tumor antigens involved in ICI therapy and the identification of related biomarkers brings us a step forward in the mechanistic understanding of ICIs and will help to monitor patients at higher risk of developing irAEs. Lastly, we discuss the current challenges in collecting research samples for the study of ICI-related mechanisms and in distinguishing between immune signatures of response and those of irAEs.
Collapse
Affiliation(s)
- Fiamma Berner
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Lukas Flatz
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
- Department of Dermatology, University Hospital Tübingen, University of Tübingen, Tübingen, Germany
- Department of Dermatology, Kantonsspital St. Gallen, St. Gallen, Switzerland
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Oncology and Hematology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| |
Collapse
|
429
|
Fortman DD, Hurd D, Davar D. The Microbiome in Advanced Melanoma: Where Are We Now? Curr Oncol Rep 2023; 25:997-1016. [PMID: 37269504 PMCID: PMC11090495 DOI: 10.1007/s11912-023-01431-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/14/2023] [Indexed: 06/05/2023]
Abstract
PURPOSE OF REVIEW This review summarizes recent data linking gut microbiota composition to ICI outcomes and gut microbiota-specific interventional clinical trials in melanoma. RECENT FINDINGS Preclinical and clinical studies have demonstrated the effects of the gut microbiome modulation upon ICI response in advanced melanoma, with growing evidence supporting the ability of the gut microbiome to restore or improve ICI response in advanced melanoma through dietary fiber, probiotics, and FMT. Immune checkpoint inhibitors (ICI) targeting the PD-1, CTLA-4, and LAG-3 negative regulatory checkpoints have transformed the management of melanoma. ICIs are FDA-approved in advanced metastatic disease, stage III resected melanoma, and high-risk stage II melanoma and are being investigated more recently in the management of high-risk resectable melanoma in the peri-operative setting. The gut microbiome has emerged as an important tumor-extrinsic modulator of both response and immune-related adverse event (irAE) development in ICI-treated cancer in general, and melanoma in particular.
Collapse
Affiliation(s)
- Dylan D Fortman
- Division of General Internal Medicine, Department of Medicine, University of Pittsburgh Medical Center (UPMC) and University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Drew Hurd
- UPMC Hillman Cancer Center, Department of Medicine, University of Pittsburgh, Pavilion, Suite 1.32d, 5115, Center Avenue, Pittsburgh, PA, 15213, USA
| | - Diwakar Davar
- UPMC Hillman Cancer Center, Department of Medicine, University of Pittsburgh, Pavilion, Suite 1.32d, 5115, Center Avenue, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
430
|
Chen S, Zhang L, Huang M, Liang Y, Wang Y. A tumor-associated endothelial signature score model in immunotherapy and prognosis across pan-cancers. Front Pharmacol 2023; 14:1190660. [PMID: 37719845 PMCID: PMC10500301 DOI: 10.3389/fphar.2023.1190660] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 08/14/2023] [Indexed: 09/19/2023] Open
Abstract
Background: The tumor-associated endothelial cell (TAE) component plays a vital role in tumor immunity. However, systematic tumor-associated endothelial-related gene assessment models for predicting cancer immunotherapy (CIT) responses and survival across human cancers have not been explored. Herein, we investigated a TAE gene risk model to predict CIT responses and patient survival in a pan-cancer analysis. Methods: We analyzed publicly available datasets of tumor samples with gene expression and clinical information, including gastric cancer, metastatic urothelial cancer, metastatic melanoma, non-small cell lung cancer, primary bladder cancer, and renal cell carcinoma. We further established a binary classification model to predict CIT responses using the least absolute shrinkage and selection operator (LASSO) computational algorithm. Results: The model demonstrated a high predictive accuracy in both training and validation cohorts. The response rate of the high score group to immunotherapy in the training cohort was significantly higher than that of the low score group, with CIT response rates of 51% and 27%, respectively. The survival analysis showed that the prognosis of the high score group was significantly better than that of the low score group (all p < 0·001). Tumor-associated endothelial gene signature scores positively correlated with immune checkpoint genes, suggesting that immune checkpoint inhibitors may benefit patients in the high score group. The analysis of TAE scores across 33 human cancers revealed that the TAE model could reflect immune cell infiltration and predict the survival of cancer patients. Conclusion: The TAE signature model could represent a CIT response prediction model with a prognostic value in multiple cancer types.
Collapse
Affiliation(s)
- Shuzhao Chen
- Department of Hematologic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Limei Zhang
- Department of Hematologic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Mayan Huang
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yang Liang
- Department of Hematologic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yun Wang
- Department of Hematologic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| |
Collapse
|
431
|
Zawidzka EM, Biavati L, Thomas A, Zanettini C, Marchionni L, Leone R, Borrello I. Tumor-Specific CD8 + T Cells from the Bone Marrow Resist Exhaustion and Exhibit Increased Persistence in Tumor-Bearing Hosts as Compared to Tumor Infiltrating Lymphocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.28.555119. [PMID: 37693379 PMCID: PMC10491133 DOI: 10.1101/2023.08.28.555119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Immunotherapy is now an integral aspect of cancer therapy. Strategies employing adoptive cell therapy (ACT) have seen the establishment of chimeric antigen receptor (CAR)-T cells using peripheral blood lymphocytes as well as tumor infiltrating lymphocytes (TILs) with significant clinical results. Despite these successes, the limitations of the current strategies are also emerging and novel approaches are needed. The bone marrow (BM) is an immunological niche that houses T cells with specificity for previously encountered antigens, including tumor-associated antigens from certain solid cancers. This study sought to improve our understanding of tumor-specific BM T cells in the context of solid tumors by comparing them with TILs, and to assess whether there is a rationale for using the BM as a source of T cells for ACT against solid malignancies. Herein, we demonstrate that T cells from the BM appear superior to TILs as a source of cells for cellular therapy. Specifically, they possess a memory-enriched phenotype and exhibit improved effector function, greater persistence within a tumor-bearing host, and the capacity for increased tumor infiltration. Taken together, these data provide a foundation for further exploring the BM as a source of tumor-specific T cells for ACT in solid malignancies.
Collapse
Affiliation(s)
- Elizabeth M. Zawidzka
- Johns Hopkins University School of Medicine, Bloomberg Kimmel Institute for Cancer Immunotherapy
| | - Luca Biavati
- Johns Hopkins University School of Medicine, Bloomberg Kimmel Institute for Cancer Immunotherapy
| | - Amy Thomas
- Johns Hopkins University School of Medicine, Bloomberg Kimmel Institute for Cancer Immunotherapy
| | | | | | - Robert Leone
- Johns Hopkins University School of Medicine, Bloomberg Kimmel Institute for Cancer Immunotherapy
| | - Ivan Borrello
- Johns Hopkins University School of Medicine, Bloomberg Kimmel Institute for Cancer Immunotherapy
- Current Address: Tampa General Hospital Cancer Institute
| |
Collapse
|
432
|
Angelova-Toshkina D, Weide B, Tietze LF, Hebst M, Tietze JK. Correlation of Baseline Tumor Burden with Clinical Outcome in Melanoma Patients Treated with Ipilimumab. Oncology 2023; 102:76-84. [PMID: 37579734 DOI: 10.1159/000533504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/03/2023] [Indexed: 08/16/2023]
Abstract
INTRODUCTION Tumor burden is a frequently mentioned parameter; however, a commonly accepted definition is still lacking. METHODS In this double-center prospective and retrospective study, 76 patients with unresectable stage III or stage IV melanoma treated with ipilimumab were included. We defined the baseline tumor burden (BTB) as the global sum of all metastases' longest diameters before treatment started and correlated the calculated BTB with disease control rate (DCR), progression-free survival (PFS), overall survival (OS), and with the baseline levels of LDH, S100B, and sULPB2. RESULTS BTB correlated significantly with DCR (p = 0.009), PFS (p = 0.002), OS (p = 0.032), and the occurrence of NRAS mutation (p = 0.006). BTB was also correlated to baseline serum levels of LDH (p = 0.011), S100B (p = 0.027), and SULBP (p < 0.0001). Multivariate analysis revealed that BPB and LDH were independently correlated with PFS and OS. With increasing BTB, disease control was less likely; no patient with a BTB >200 mm achieved disease control. For patients with brain metastasis, no correlation of BTB with DCR (p = 0.251), PFS (p = 0.059), or OS (p = 0.981) was observed. CONCLUSION Calculated BTB is an independent prognostic factor for patients with metastatic melanoma treated with ipilimumab. Using calculated BTB as a definition of tumor burden may help increase comparability of outcome of therapies in future studies.
Collapse
Affiliation(s)
| | - Benjamin Weide
- Department of Dermatology, University Hospital Tübingen, Tübingen, Germany
| | - Lutz F Tietze
- Institute of Organic and Biomolecular Chemistry, Georg-August University, Göttingen, Germany
| | - Michelle Hebst
- Clinic and Policlinic for Dermatology and Venereology, University Medical Center Rostock, Rostock, Germany
| | - Julia K Tietze
- Clinic and Policlinic for Dermatology and Venereology, University Medical Center Rostock, Rostock, Germany
| |
Collapse
|
433
|
Sandbank E, Eckerling A, Margalit A, Sorski L, Ben-Eliyahu S. Immunotherapy during the Immediate Perioperative Period: A Promising Approach against Metastatic Disease. Curr Oncol 2023; 30:7450-7477. [PMID: 37623021 PMCID: PMC10453707 DOI: 10.3390/curroncol30080540] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/26/2023] Open
Abstract
Tumor excision is a necessary life-saving procedure in most solid cancers. However, surgery and the days before and following it, known as the immediate perioperative period (IPP), entail numerous prometastatic processes, including the suppression of antimetastatic immunity and direct stimulation of minimal residual disease (MRD). Thus, the IPP is pivotal in determining long-term cancer outcomes, presenting a short window of opportunity to circumvent perioperative risk factors by employing several therapeutic approaches, including immunotherapy. Nevertheless, immunotherapy is rarely examined or implemented during this short timeframe, due to both established and hypothetical contraindications to surgery. Herein, we analyze how various aspects of the IPP promote immunosuppression and progression of MRD, and how potential IPP application of immunotherapy may interact with these deleterious processes. We discuss the feasibility and safety of different immunotherapies during the IPP with a focus on the latest approaches of immune checkpoint inhibition. Last, we address the few past and ongoing clinical trials that exploit the IPP timeframe for anticancer immunotherapy. Accordingly, we suggest that several specific immunotherapies can be safely and successfully applied during the IPP, alone or with supporting interventions, which may improve patients' resistance to MRD and overall survival.
Collapse
Affiliation(s)
- Elad Sandbank
- Neuro-Immunology Research Unit, School of Psychological Sciences, Tel Aviv University, Tel Aviv 69978, Israel; (E.S.); (A.E.); (L.S.)
| | - Anabel Eckerling
- Neuro-Immunology Research Unit, School of Psychological Sciences, Tel Aviv University, Tel Aviv 69978, Israel; (E.S.); (A.E.); (L.S.)
| | - Adam Margalit
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel;
| | - Liat Sorski
- Neuro-Immunology Research Unit, School of Psychological Sciences, Tel Aviv University, Tel Aviv 69978, Israel; (E.S.); (A.E.); (L.S.)
| | - Shamgar Ben-Eliyahu
- Neuro-Immunology Research Unit, School of Psychological Sciences, Tel Aviv University, Tel Aviv 69978, Israel; (E.S.); (A.E.); (L.S.)
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel;
| |
Collapse
|
434
|
Profili NI, Castelli R, Gidaro A, Merella A, Manetti R, Palmieri G, Maioli M, Delitala AP. Endocrine Side Effects in Patients Treated with Immune Checkpoint Inhibitors: A Narrative Review. J Clin Med 2023; 12:5161. [PMID: 37568563 PMCID: PMC10419837 DOI: 10.3390/jcm12155161] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/19/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023] Open
Abstract
Checkpoint inhibitors are monoclonal antibodies that elicit an anti-tumor response by stimulating immune system. Their use has improved the treatment of different types of cancer such as melanoma, breast carcinoma, lung, stomach, colon, liver, renal cell carcinoma, and Hodgkin's lymphoma, but several adverse events have been reported. Although the etiology of these effects is not completely understood, an uncontrolled activation of the immune system has been postulated. Indeed, some studies showed a cross reactivity of T cells, which acted against tumor antigens as well as antigens in the tissues of patients who developed immune-related adverse events. Despite the known possibility of developing immune-related adverse events, early diagnosis, monitoring during therapy, and treatment are fundamental for the best supportive care and administration of immune checkpoint inhibitors. The aim of this review is to guide the clinician in early diagnosis, management, and treatment of the endocrinological adverse effects in the major endocrine glands (thyroid, pituitary, adrenal, endocrine pancreas, and parathyroid).
Collapse
Affiliation(s)
- Nicia I. Profili
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy
| | - Roberto Castelli
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy
| | - Antonio Gidaro
- Department of Biomedical and Clinical Sciences Luigi Sacco, University of Milan, Luigi Sacco Hospital, 20157 Milan, Italy
| | - Alessandro Merella
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy
| | - Roberto Manetti
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy
| | - Giuseppe Palmieri
- Department of Biochemical Science, University of Sassari, 07100 Sassari, Italy
| | - Margherita Maioli
- Department of Biochemical Science, University of Sassari, 07100 Sassari, Italy
| | - Alessandro P. Delitala
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy
| |
Collapse
|
435
|
Rizzetto G, De Simoni E, Molinelli E, Offidani A, Simonetti O. Efficacy of Pembrolizumab in Advanced Melanoma: A Narrative Review. Int J Mol Sci 2023; 24:12383. [PMID: 37569757 PMCID: PMC10419154 DOI: 10.3390/ijms241512383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/29/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
Pembrolizumab has been shown to increase survival in patients with metastatic melanoma. Considering the numerous oncoming studies, we decided to conduct a narrative review of the latest efficacy evidence regarding the use of pembrolizumab, alone or in combination, in patients with metastatic melanoma. A search was conducted in PubMed using "pembrolizumab," and "metastatic melanoma" as keywords, considering studies from 2022 onward. We reviewed pembrolizumab and associations, cost-effectiveness, virus, advanced acral melanoma, long-term outcomes, real-life data, biomarkers, obesity, and vaccines. In conclusion, pembrolizumab is a fundamental option in the therapy of metastatic melanoma. However, a certain group of patients do not respond and, therefore, new combination options need to be evaluated. In particular, the use of vaccines tailored to tumor epitopes could represent a breakthrough in the treatment of resistant forms. Further studies with larger sample numbers are needed to confirm the preliminary results.
Collapse
Affiliation(s)
| | | | | | | | - Oriana Simonetti
- Clinic of Dermatology, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, 60126 Ancona, Italy; (G.R.); (E.D.S.); (E.M.); (A.O.)
| |
Collapse
|
436
|
Nardo M, Yilmaz B, Nelson BE, Torres HA, Wang LS, Granwehr BP, Song J, Dalla Pria HRF, Trinh VA, Glitza Oliva IC, Patel SP, Tannir NM, Kaseb AO, Altan M, Lee SS, Miller E, Zhang H, Stephen BA, Naing A. Safety and Efficacy of Immune Checkpoint Inhibitors in Patients with Cancer and Viral Hepatitis: The MD Anderson Cancer Center Experience. Oncologist 2023; 28:714-721. [PMID: 36952233 PMCID: PMC10400154 DOI: 10.1093/oncolo/oyad039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 01/24/2023] [Indexed: 03/24/2023] Open
Abstract
BACKGROUND Despite the clinical benefit of immune checkpoint inhibitors (ICIs), patients with a viral hepatitis have been excluded from clinical trials because of safety concerns. The purpose of this study was to determine the incidence rate of adverse events (AEs) in patients with viral hepatitis who received ICIs for cancer treatment. MATERIALS AND METHODS We conducted a retrospective study in patients with cancer and concurrent hepatitis B or C, who had undergone treatment with ICI at MD Anderson Cancer Center from January 1, 2010 to December 31, 2019. RESULTS Of the 1076 patients screened, we identified 33 with concurrent hepatitis. All 10 patients with HBV underwent concomitant antiviral therapy during ICI treatment. Sixteen of the 23 patients with HCV received it before the initiation of ICI. The median follow-up time was 33 months (95% CI, 23-45) and the median duration of ICI therapy was 3 months (IQR, 1.9-6.6). Of the 33 patients, 12 (39%) experienced irAEs (immune-related adverse events) of any grade, with 2 (6%) having grade 3 or higher. None of the patients developed hepatitis toxicities. CONCLUSION ICIs may be a therapeutic option with an acceptable safety profile in patients with cancer and advanced liver disease.
Collapse
Affiliation(s)
- Mirella Nardo
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bulent Yilmaz
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Blessie Elizabeth Nelson
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Harrys A Torres
- Department of Infectious Diseases Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lan Sun Wang
- Department of Genitourinary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bruno Palma Granwehr
- Department of Infectious Diseases Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Juhee Song
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hanna R F Dalla Pria
- Department of Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Van A Trinh
- Department of Melanoma Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Isabella C Glitza Oliva
- Department of Melanoma Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sapna P Patel
- Department of Melanoma Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nizar M Tannir
- Department of Genitourinary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ahmed Omar Kaseb
- Department of Genitourinary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mehmet Altan
- Department of Genitourinary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston TX, USA
| | - Sunyoung S Lee
- Department of Gastrointestinal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ethan Miller
- Department of Gastrointestinal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hao Zhang
- Department of Gastrointestinal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bettzy A Stephen
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Aung Naing
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
437
|
Wan PKT, Fernandes RA, Seymour LW. Oncolytic viruses and antibodies: are they more successful when delivered separately or when engineered as a single agent? J Immunother Cancer 2023; 11:e006518. [PMID: 37541690 PMCID: PMC10407364 DOI: 10.1136/jitc-2022-006518] [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] [Accepted: 06/25/2023] [Indexed: 08/06/2023] Open
Abstract
Oncolytic viruses (OVs) provide the promise of tumor-selective cytotoxicity coupled with amplification of the therapeutic agent (the virus) in situ within the tumor improving its therapeutic index. Despite this promise, however, single agent-treatments have not been as successful as combination therapies, particularly combining with checkpoint inhibitor antibodies. The antibodies may be delivered by two approaches, either encoded within the OV genome to restrict antibody production to sites of active virus infection or alternatively given alongside OVs as separate treatments. Both approaches have shown promising therapeutic outcomes, and this leads to an interesting question of whether one approach is potentially better than the other. In this review, we provide a brief summary of the combination OV-antibody therapies that target tumor cells, tumor microenvironment and immune cells to help define key parameters influencing which approach is superior, thereby improving insight into the rational design of OV treatment strategies.
Collapse
|
438
|
Routy B, Lenehan JG, Miller WH, Jamal R, Messaoudene M, Daisley BA, Hes C, Al KF, Martinez-Gili L, Punčochář M, Ernst S, Logan D, Belanger K, Esfahani K, Richard C, Ninkov M, Piccinno G, Armanini F, Pinto F, Krishnamoorthy M, Figueredo R, Thebault P, Takis P, Magrill J, Ramsay L, Derosa L, Marchesi JR, Parvathy SN, Elkrief A, Watson IR, Lapointe R, Segata N, Haeryfar SMM, Mullish BH, Silverman MS, Burton JP, Maleki Vareki S. Fecal microbiota transplantation plus anti-PD-1 immunotherapy in advanced melanoma: a phase I trial. Nat Med 2023; 29:2121-2132. [PMID: 37414899 DOI: 10.1038/s41591-023-02453-x] [Citation(s) in RCA: 198] [Impact Index Per Article: 99.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 06/08/2023] [Indexed: 07/08/2023]
Abstract
Fecal microbiota transplantation (FMT) represents a potential strategy to overcome resistance to immune checkpoint inhibitors in patients with refractory melanoma; however, the role of FMT in first-line treatment settings has not been evaluated. We conducted a multicenter phase I trial combining healthy donor FMT with the PD-1 inhibitors nivolumab or pembrolizumab in 20 previously untreated patients with advanced melanoma. The primary end point was safety. No grade 3 adverse events were reported from FMT alone. Five patients (25%) experienced grade 3 immune-related adverse events from combination therapy. Key secondary end points were objective response rate, changes in gut microbiome composition and systemic immune and metabolomics analyses. The objective response rate was 65% (13 of 20), including four (20%) complete responses. Longitudinal microbiome profiling revealed that all patients engrafted strains from their respective donors; however, the acquired similarity between donor and patient microbiomes only increased over time in responders. Responders experienced an enrichment of immunogenic and a loss of deleterious bacteria following FMT. Avatar mouse models confirmed the role of healthy donor feces in increasing anti-PD-1 efficacy. Our results show that FMT from healthy donors is safe in the first-line setting and warrants further investigation in combination with immune checkpoint inhibitors. ClinicalTrials.gov identifier NCT03772899 .
Collapse
Affiliation(s)
- Bertrand Routy
- Research Center of the Centre Hospitalier de l'Université de Montréal, Montréal (CRCHUM), Montreal, Quebec, Canada
- Hematology-Oncology Division, Department of Medicine, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada
| | - John G Lenehan
- Department of Oncology, Western University, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
| | - Wilson H Miller
- Lady Davis Institute of the Jewish General Hospital, Segal Cancer Centre, Montreal, Quebec, Canada
- Departments of Oncology and Medicine, McGill University, Montreal, Quebec, Canada
| | - Rahima Jamal
- Research Center of the Centre Hospitalier de l'Université de Montréal, Montréal (CRCHUM), Montreal, Quebec, Canada
- Hematology-Oncology Division, Department of Medicine, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada
| | - Meriem Messaoudene
- Research Center of the Centre Hospitalier de l'Université de Montréal, Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Brendan A Daisley
- Department of Microbiology & Immunology, Western University, London, Ontario, Canada
- Canadian Centre for Human Microbiome and Probiotics Research, London, Ontario, Canada
| | - Cecilia Hes
- Research Center of the Centre Hospitalier de l'Université de Montréal, Montréal (CRCHUM), Montreal, Quebec, Canada
- Peter Brojde Lung Cancer Center, Jewish General Hospital, Montreal, Quebec, Canada
| | - Kait F Al
- Lawson Health Research Institute, London, Ontario, Canada
- Department of Microbiology & Immunology, Western University, London, Ontario, Canada
- Canadian Centre for Human Microbiome and Probiotics Research, London, Ontario, Canada
| | - Laura Martinez-Gili
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, St Mary's Hospital Campus, Imperial College London, London, UK
- Section of Bioinformatics, Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
| | - Michal Punčochář
- Department of Computational, Cellular and Integrative Biology, University of Trento, Trento, Italy
| | - Scott Ernst
- Department of Oncology, Western University, London, Ontario, Canada
| | - Diane Logan
- Department of Oncology, Western University, London, Ontario, Canada
| | - Karl Belanger
- Research Center of the Centre Hospitalier de l'Université de Montréal, Montréal (CRCHUM), Montreal, Quebec, Canada
- Hematology-Oncology Division, Department of Medicine, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada
| | - Khashayar Esfahani
- Lady Davis Institute of the Jewish General Hospital, Segal Cancer Centre, Montreal, Quebec, Canada
- Departments of Oncology and Medicine, McGill University, Montreal, Quebec, Canada
| | - Corentin Richard
- Research Center of the Centre Hospitalier de l'Université de Montréal, Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Marina Ninkov
- Department of Microbiology & Immunology, Western University, London, Ontario, Canada
| | - Gianmarco Piccinno
- Department of Computational, Cellular and Integrative Biology, University of Trento, Trento, Italy
| | - Federica Armanini
- Department of Computational, Cellular and Integrative Biology, University of Trento, Trento, Italy
| | - Federica Pinto
- Department of Computational, Cellular and Integrative Biology, University of Trento, Trento, Italy
| | - Mithunah Krishnamoorthy
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| | - Rene Figueredo
- Department of Oncology, Western University, London, Ontario, Canada
| | - Pamela Thebault
- Research Center of the Centre Hospitalier de l'Université de Montréal, Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Panteleimon Takis
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, National Phenome Centre, Imperial College London, London, UK
- Section of Bioanalytical Chemistry, Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Jamie Magrill
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, Quebec, Canada
| | - LeeAnn Ramsay
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, Quebec, Canada
| | - Lisa Derosa
- Gustave Roussy Cancer Campus, Villejuif, France
- Cancer Medicine Department, Gustave Roussy, Villejuif, France
- Institut National de la Santé Et et de la Recherche Médicale (INSERM) U1015, ClinicObiome, Equipe Labellisée-28 Ligue Nationale contre le Cancer, Villejuif, France
- Université Paris-Saclay, Ile-de-France, France
| | - Julian R Marchesi
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, St Mary's Hospital Campus, Imperial College London, London, UK
| | - Seema Nair Parvathy
- Department of Medicine, Division of Infectious Diseases, Western University, London, Ontario, Canada
- Division of Infectious Diseases, St Joseph's Health Care, London, Ontario, Canada
| | - Arielle Elkrief
- Research Center of the Centre Hospitalier de l'Université de Montréal, Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Ian R Watson
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, Quebec, Canada
- Department of Biochemistry, McGill University, Montréal, Quebec, Canada
| | - Rejean Lapointe
- Research Center of the Centre Hospitalier de l'Université de Montréal, Montréal (CRCHUM), Montreal, Quebec, Canada
- Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, Quebec, Canada
| | - Nicola Segata
- Department of Computational, Cellular and Integrative Biology, University of Trento, Trento, Italy
| | - S M Mansour Haeryfar
- Lawson Health Research Institute, London, Ontario, Canada
- Department of Microbiology & Immunology, Western University, London, Ontario, Canada
- Department of Medicine, Division of Clinical Immunology and Allergy, Western University, London, Ontario, Canada
- Department of Surgery, Division of General Surgery, Western University, London, Ontario, Canada
| | - Benjamin H Mullish
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, St Mary's Hospital Campus, Imperial College London, London, UK
- Departments of Gastroenterology and Hepatology, St Mary's Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Michael S Silverman
- Lawson Health Research Institute, London, Ontario, Canada
- Department of Microbiology & Immunology, Western University, London, Ontario, Canada
- Department of Medicine, Division of Infectious Diseases, Western University, London, Ontario, Canada
- Division of Infectious Diseases, St Joseph's Health Care, London, Ontario, Canada
| | - Jeremy P Burton
- Lawson Health Research Institute, London, Ontario, Canada
- Department of Microbiology & Immunology, Western University, London, Ontario, Canada
- Canadian Centre for Human Microbiome and Probiotics Research, London, Ontario, Canada
| | - Saman Maleki Vareki
- Department of Oncology, Western University, London, Ontario, Canada.
- Lawson Health Research Institute, London, Ontario, Canada.
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada.
- Department of Medical Biophysics, Western University, London, Ontario, Canada.
- Ontario Institute of Cancer Research, Toronto, Ontario, Canada.
| |
Collapse
|
439
|
Villani A, Scalvenzi M, Micali G, Lacarrubba F, Fornaro L, Martora F, Potestio L. Management of Advanced Invasive Melanoma: New Strategies. Adv Ther 2023; 40:3381-3394. [PMID: 37306810 PMCID: PMC10329960 DOI: 10.1007/s12325-023-02555-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 05/15/2023] [Indexed: 06/13/2023]
Abstract
The incidence of cutaneous melanoma (CM) is increasing. CM is defined as melanoma in situ when limited within the epidermis and invasive when atypical melanocytes progressively invade the dermis. Treatment of CM is challenging. On one hand, melanoma in situ does not require further treatment except for a limited secondary excision with reduced margins to minimize the risk of local recurrences; on the other, invasive melanoma requires a personalized approach based on tumor staging. Consequently, an association of surgical and medical treatments is often necessary for invasive forms of the disease. In this scenario, new knowledge on melanoma pathogenesis has led to the development of safe and effective treatments, and several drugs are currently under investigation. However, extensive knowledge is required to offer patients a tailored-tail approach. The aim of our article was to review current literature to provide an overview of treatment options for invasive melanoma, highlighting strategical approaches that can be used in patients with these forms of disease.
Collapse
Affiliation(s)
- Alessia Villani
- Dermatology Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, Via Pansini, 5, 80131, Naples, Italy.
| | - Massimiliano Scalvenzi
- Dermatology Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, Via Pansini, 5, 80131, Naples, Italy
| | | | | | - Luigi Fornaro
- Dermatology Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, Via Pansini, 5, 80131, Naples, Italy
| | - Fabrizio Martora
- Dermatology Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, Via Pansini, 5, 80131, Naples, Italy
| | - Luca Potestio
- Dermatology Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, Via Pansini, 5, 80131, Naples, Italy
| |
Collapse
|
440
|
Ferguson R, Chat V, Morales L, Simpson D, Monson KR, Cohen E, Zusin S, Madonna G, Capone M, Simeone E, Pavlick A, Luke JJ, Gajewski TF, Osman I, Ascierto P, Weber J, Kirchhoff T. Germline immunomodulatory expression quantitative trait loci (ieQTLs) associated with immune-related toxicity from checkpoint inhibition. Eur J Cancer 2023; 189:112923. [PMID: 37301715 PMCID: PMC11000635 DOI: 10.1016/j.ejca.2023.05.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/11/2023] [Accepted: 05/13/2023] [Indexed: 06/12/2023]
Abstract
BACKGROUND Immune checkpoint inhibition (ICI) has improved clinical outcomes for metastatic melanoma patients; however, 65-80% of patients treated with ICI experience immune-related adverse events (irAEs). Given the plausible link of irAEs with underlying host immunity, we explored whether germline genetic variants controlling the expression of 42 immunomodulatory genes were associated with the risk of irAEs in melanoma patients treated with the single-agent anti-CTLA-4 antibody ipilimumab (IPI). METHODS We identified 42 immunomodulatory expression quantitative trait loci (ieQTLs) most significantly associated with the expression of 382 immune-related genes. These germline variants were genotyped in IPI-treated melanoma patients, collected as part of a multi-institutional collaboration. We tested the association of ieQTLs with irAEs in a discovery cohort of 95 patients, followed by validation in an additional 97 patients. RESULTS We found that the alternate allele of rs7036417, a variant linked to increased expression of SYK, was strongly associated with an increased risk of grade 3-4 toxicity [odds ratio (OR) = 7.46; 95% confidence interval (CI) = 2.65-21.03; p = 1.43E-04]. This variant was not associated with response (OR = 0.90; 95% CI = 0.37-2.21; p = 0.82). CONCLUSION We report that rs7036417 is associated with increased risk of severe irAEs, independent of IPI efficacy. SYK plays an important role in B-cell/T-cell expansion, and increased pSYK has been reported in patients with autoimmune disease. The association between rs7036417 and IPI irAEs in our data suggests a role of SYK overexpression in irAE development. These findings support the hypothesis that inherited variation in immune-related pathways modulates ICI toxicity and suggests SYK as a possible future target for therapies to reduce irAEs.
Collapse
Affiliation(s)
- Robert Ferguson
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA; Departments of Population Health and Environmental Medicine, New York University-Grossman School of Medicine, New York, NY, USA; The Interdisciplinary Melanoma Cooperative Group, New York University-Grossman School of Medicine, New York, NY, USA
| | - Vylyny Chat
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA; Departments of Population Health and Environmental Medicine, New York University-Grossman School of Medicine, New York, NY, USA; The Interdisciplinary Melanoma Cooperative Group, New York University-Grossman School of Medicine, New York, NY, USA
| | - Leah Morales
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA; Departments of Population Health and Environmental Medicine, New York University-Grossman School of Medicine, New York, NY, USA; The Interdisciplinary Melanoma Cooperative Group, New York University-Grossman School of Medicine, New York, NY, USA
| | - Danny Simpson
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA; Departments of Population Health and Environmental Medicine, New York University-Grossman School of Medicine, New York, NY, USA; The Interdisciplinary Melanoma Cooperative Group, New York University-Grossman School of Medicine, New York, NY, USA
| | - Kelsey R Monson
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA; Departments of Population Health and Environmental Medicine, New York University-Grossman School of Medicine, New York, NY, USA; The Interdisciplinary Melanoma Cooperative Group, New York University-Grossman School of Medicine, New York, NY, USA
| | - Elisheva Cohen
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA; Departments of Population Health and Environmental Medicine, New York University-Grossman School of Medicine, New York, NY, USA; The Interdisciplinary Melanoma Cooperative Group, New York University-Grossman School of Medicine, New York, NY, USA
| | - Sarah Zusin
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA; Departments of Population Health and Environmental Medicine, New York University-Grossman School of Medicine, New York, NY, USA; The Interdisciplinary Melanoma Cooperative Group, New York University-Grossman School of Medicine, New York, NY, USA
| | - Gabriele Madonna
- Melanoma Cancer Immunotherapy and Innovative Therapy Unit, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Napoli, Italy
| | - Mariaelena Capone
- Melanoma Cancer Immunotherapy and Innovative Therapy Unit, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Napoli, Italy
| | - Ester Simeone
- Melanoma Cancer Immunotherapy and Innovative Therapy Unit, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Napoli, Italy
| | - Anna Pavlick
- Division of Hematology & Medical Oncology, the Cutaneous Oncology Program, Weill Cornell Medicine and New York-Presbyterian, New York, USA
| | - Jason J Luke
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA; UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - Thomas F Gajewski
- Department of Pathology, University of Chicago, Chicago, IL, USA; Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA; Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Iman Osman
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA; The Interdisciplinary Melanoma Cooperative Group, New York University-Grossman School of Medicine, New York, NY, USA; Department of Medicine, New York University-Grossman School of Medicine, New York, NY, USA; Ronald O. Perelman Department of Dermatology, New York University-Grossman School of Medicine, New York, NY, USA
| | - Paolo Ascierto
- Melanoma Cancer Immunotherapy and Innovative Therapy Unit, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Napoli, Italy
| | - Jeffrey Weber
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA; The Interdisciplinary Melanoma Cooperative Group, New York University-Grossman School of Medicine, New York, NY, USA; Department of Medicine, New York University-Grossman School of Medicine, New York, NY, USA
| | - Tomas Kirchhoff
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA; Departments of Population Health and Environmental Medicine, New York University-Grossman School of Medicine, New York, NY, USA; The Interdisciplinary Melanoma Cooperative Group, New York University-Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
441
|
Herpels M, Ishihara J, Sadanandam A. The clinical terrain of immunotherapies in heterogeneous pancreatic cancer: unravelling challenges and opportunities. J Pathol 2023; 260:533-550. [PMID: 37550956 DOI: 10.1002/path.6171] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/20/2023] [Accepted: 06/22/2023] [Indexed: 08/09/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most common and aggressive type of pancreatic cancer and has abysmal survival rates. In the past two decades, immunotherapeutic agents with success in other cancer types have gradually been trialled against PDACs at different stages of cancer progression, either as a monotherapy or in combination with chemotherapy. Unfortunately, to this day, chemotherapy still prolongs the survival rates the most and is prescribed in clinics despite the severe side effects in other cancer types. The low success rates of immunotherapy against PDAC have been attributed most frequently to its complex and multi-faceted tumour microenvironment (TME) and low mutational burden. In this review, we give a comprehensive overview of the immunotherapies tested in PDAC clinical trials thus far, their limitations, and potential explanations for their failure. We also discuss the existing classification of heterogenous PDACs into cancer, cancer-associated fibroblast, and immune subtypes and their potential opportunity in patient selection as a form of personalisation of PDAC immunotherapy. © 2023 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Melanie Herpels
- Division of Molecular Pathology, Institute of Cancer Research, London, UK
- Department of Bioengineering, Imperial College London, London, UK
| | - Jun Ishihara
- Department of Bioengineering, Imperial College London, London, UK
| | - Anguraj Sadanandam
- Division of Molecular Pathology, Institute of Cancer Research, London, UK
- Centre for Global Oncology, Division of Molecular Pathology, Institute of Cancer Research, London, UK
- Centre for Translational Immunotherapy, Division of Radiotherapy and Imaging, Institute of Cancer Research, London, UK
| |
Collapse
|
442
|
Fröhlich F, Ramelyte E, Turko P, Dzung A, Freiberger SN, Mangana J, Levesque MP, Dummer R. Clock-like Mutation Signature May Be Prognostic for Worse Survival Than Signatures of UV Damage in Cutaneous Melanoma. Cancers (Basel) 2023; 15:3818. [PMID: 37568633 PMCID: PMC10418148 DOI: 10.3390/cancers15153818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/14/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Novel treatment modalities comprising immune checkpoint inhibitors and targeted therapies have revolutionized treatment of metastatic melanoma. Still, some patients suffer from rapid progression and decease within months after a diagnosis of stage IV melanoma. We aimed to assess whether genomic alterations may predict survival after the development of stage IV disease, irrespective of received therapy. We analyzed tumor samples of 79 patients with stage IV melanoma using a custom next-generation gene-sequencing panel, MelArray, designed to detect alterations in 190 melanoma-relevant genes. We classified the patients: first, as short survivors (survival ≤6 months after stage IV disease, n = 22) and long survivors (survival >6 months, n = 57); second, by using a cut-off of one year; and third, by comparing the longest surviving 20 patients to the shortest surviving 20. Among analyzed genes, no individual gene alterations, or combinations of alterations, could be dichotomously associated with survival. However, the cohort's mutational profiles closely matched three known mutational signatures curated by the Catalog of Somatic Mutations in Cancer (COSMIC): UV signature COSMIC_7 (cosine-similarity 0.932), clock-like signature COSMIC_5 (cosine-similarity 0.829), and COSMIC_30 (cosine-similarity 0.726). Patients with UV signature had longer survival compared to patients with clock-like and COSMIC 30 (p < 0.0001). Subgroup dichotomization at 6 months showed that 75% of patients with UV signature survived longer than 6 months, and about 75% of patients with clock-like signature survived less than 6 months after development of stage IV disease. In our cohort, clock-like COSMIC_5 mutational signature predicted poor survival while a UV signature COSMIC_7 predicted longer survival. The prognostic value of mutational signatures should be evaluated in prospective studies.
Collapse
Affiliation(s)
- Fabienne Fröhlich
- Department of Dermatology, University Hospital of Zurich, 8091 Zurich, Switzerland; (F.F.); (E.R.); (P.T.); (A.D.); (J.M.); (M.P.L.)
- Faculty of Medicine, University of Zurich, 8091 Zurich, Switzerland
| | - Egle Ramelyte
- Department of Dermatology, University Hospital of Zurich, 8091 Zurich, Switzerland; (F.F.); (E.R.); (P.T.); (A.D.); (J.M.); (M.P.L.)
- Faculty of Medicine, University of Zurich, 8091 Zurich, Switzerland
| | - Patrick Turko
- Department of Dermatology, University Hospital of Zurich, 8091 Zurich, Switzerland; (F.F.); (E.R.); (P.T.); (A.D.); (J.M.); (M.P.L.)
- Faculty of Medicine, University of Zurich, 8091 Zurich, Switzerland
| | - Andreas Dzung
- Department of Dermatology, University Hospital of Zurich, 8091 Zurich, Switzerland; (F.F.); (E.R.); (P.T.); (A.D.); (J.M.); (M.P.L.)
- Faculty of Medicine, University of Zurich, 8091 Zurich, Switzerland
| | - Sandra N. Freiberger
- Department of Pathology, University Hospital of Zurich, 8091 Zurich, Switzerland;
| | - Joanna Mangana
- Department of Dermatology, University Hospital of Zurich, 8091 Zurich, Switzerland; (F.F.); (E.R.); (P.T.); (A.D.); (J.M.); (M.P.L.)
- Faculty of Medicine, University of Zurich, 8091 Zurich, Switzerland
| | - Mitchell P. Levesque
- Department of Dermatology, University Hospital of Zurich, 8091 Zurich, Switzerland; (F.F.); (E.R.); (P.T.); (A.D.); (J.M.); (M.P.L.)
- Faculty of Medicine, University of Zurich, 8091 Zurich, Switzerland
| | - Reinhard Dummer
- Department of Dermatology, University Hospital of Zurich, 8091 Zurich, Switzerland; (F.F.); (E.R.); (P.T.); (A.D.); (J.M.); (M.P.L.)
- Faculty of Medicine, University of Zurich, 8091 Zurich, Switzerland
| |
Collapse
|
443
|
Tang W, Chen J, Ji T, Cong X. TIGIT, a novel immune checkpoint therapy for melanoma. Cell Death Dis 2023; 14:466. [PMID: 37495610 PMCID: PMC10372028 DOI: 10.1038/s41419-023-05961-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 06/20/2023] [Accepted: 07/05/2023] [Indexed: 07/28/2023]
Abstract
Melanoma is the most aggressive and deadliest type of skin cancer. In the last 10 years, immune checkpoint blockades (ICBs) including PD-1/PD-L1 and CTLA-4 inhibitor has been shown to be effective against melanoma. PD-1/PD-L1 and CTLA-4 inhibitors have shown varying degrees of drug resistance in the treatment of melanoma patients. Furthermore, the clinical benefits of ICBs are also accompanied by severe immune toxicity. Therefore, there is an urgent need to develop new immune checkpoint inhibitors to optimize melanoma therapy and reduce cytotoxicity. T-cell immunoreceptor with immunoglobulin and immunoreceptor tyrosine-based inhibition motif domain (TIGIT) is thought to activate inhibitory receptors in T cells, natural killer (NK) cells, and regulatory T cells (Tregs), and has become a promising target for immunotherapy. Studies have found that TIGIT can be detected in different stages of melanoma, which is closely related to the occurrence, development, and prognosis of melanoma. This review mainly describes the immunosuppressive mechanism of TIGIT and its role in antitumor immunity of melanoma, so as to provide new ideas and schemes for the clinical treatment of melanoma with targeted TIGIT.
Collapse
Affiliation(s)
- Wei Tang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110000, Liaoning Province, China
| | - Jun Chen
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110000, Liaoning Province, China
| | - Tianlong Ji
- Department of Radiation Oncology, The First Hospital of China Medical University, Shenyang, 110000, China.
| | - Xiufeng Cong
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110000, Liaoning Province, China.
| |
Collapse
|
444
|
Sun L, Shen F, Qu Y, Liu Z. Functional DNA as a Molecular Tool in Regulating Immunoreceptor-Ligand Interactions. JACS AU 2023; 3:1820-1834. [PMID: 37502159 PMCID: PMC10369416 DOI: 10.1021/jacsau.3c00291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 06/24/2023] [Accepted: 06/27/2023] [Indexed: 07/29/2023]
Abstract
During immune responses, activating ligands would trigger dynamic spatiotemporal organization of immunoreceptors at the cell interface, governing the fate and effector functions of immune cells. To understand the biophysical mechanisms of immunoreceptor signaling, diverse tools, including DNA technologies, have been developed to manipulate receptor-ligand interactions during the immune activation process. With great capability in the controllable assembly of biomolecules, functional DNA-based precise arrangement of immune molecules at cell interfaces has provided a powerful means in revealing the principles of immunoreceptor triggering, even at the single-molecule level. In addition, precisely regulating immunoreceptor-ligand interactions with functional DNA has been applied in immunotherapies of major diseases. This Perspective will focus on the recent advances in exploring immunoreceptor signaling with functional DNA as the molecular tool as well as the applications of functional DNA mediated regulation of immunoreceptor activation. We also outline the challenges and opportunities of applying functional DNA in immune modulation and immunotherapy.
Collapse
Affiliation(s)
- Lele Sun
- Institute
of Materiobiology, Department of Chemistry, College of Science, Shanghai University, Shanghai 200444, China
| | - Fengyun Shen
- School
of Chemistry and Chemical Engineering, Shanghai
Jiao Tong University, Shanghai 201240, China
| | - Yanfei Qu
- Institute
of Materiobiology, Department of Chemistry, College of Science, Shanghai University, Shanghai 200444, China
| | - Zhuang Liu
- Institute
of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Lab
Carbon Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| |
Collapse
|
445
|
Long GV, Swetter SM, Menzies AM, Gershenwald JE, Scolyer RA. Cutaneous melanoma. Lancet 2023:S0140-6736(23)00821-8. [PMID: 37499671 DOI: 10.1016/s0140-6736(23)00821-8] [Citation(s) in RCA: 274] [Impact Index Per Article: 137.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 03/17/2023] [Accepted: 04/19/2023] [Indexed: 07/29/2023]
Abstract
Cutaneous melanoma is a malignancy arising from melanocytes of the skin. Incidence rates are rising, particularly in White populations. Cutaneous melanoma is typically driven by exposure to ultraviolet radiation from natural sunlight and indoor tanning, although there are several subtypes that are not related to ultraviolet radiation exposure. Primary melanomas are often darkly pigmented, but can be amelanotic, with diagnosis based on a combination of clinical and histopathological findings. Primary melanoma is treated with wide excision, with margins determined by tumour thickness. Further treatment depends on the disease stage (following histopathological examination and, where appropriate, sentinel lymph node biopsy) and can include surgery, checkpoint immunotherapy, targeted therapy, or radiotherapy. Systemic drug therapies are recommended as an adjunct to surgery in patients with resectable locoregional metastases and are the mainstay of treatment in advanced melanoma. Management of advanced melanoma is complex, particularly in those with cerebral metastasis. Multidisciplinary care is essential. Systemic drug therapies, particularly immune checkpoint inhibitors, have substantially increased melanoma survival following a series of landmark approvals from 2011 onward.
Collapse
Affiliation(s)
- Georgina V Long
- Melanoma Institute Australia, Sydney, NSW, Australia; Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia; Department of Medical Oncology, Royal North Shore Hospital, Sydney, NSW, Australia; Department of Medical Oncology, Mater Hospital, Sydney, NSW, Australia.
| | - Susan M Swetter
- Department of Dermatology and Pigmented Lesion and Melanoma Program, Stanford University Medical Center and Cancer Institute, Stanford, CA, USA; Department of Dermatology, VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Alexander M Menzies
- Melanoma Institute Australia, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia; Department of Medical Oncology, Royal North Shore Hospital, Sydney, NSW, Australia; Department of Medical Oncology, Mater Hospital, Sydney, NSW, Australia
| | - Jeffrey E Gershenwald
- Department of Surgical Oncology and Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Richard A Scolyer
- Melanoma Institute Australia, Sydney, NSW, Australia; Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia; Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Sydney, NSW, Australia; NSW Health Pathology, Sydney, NSW, Australia
| |
Collapse
|
446
|
Wang Y, Zhong X, He X, Hu Z, Huang H, Chen J, Chen K, Zhao S, Wei P, Li D. Liver metastasis from colorectal cancer: pathogenetic development, immune landscape of the tumour microenvironment and therapeutic approaches. J Exp Clin Cancer Res 2023; 42:177. [PMID: 37480104 PMCID: PMC10362774 DOI: 10.1186/s13046-023-02729-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 06/05/2023] [Indexed: 07/23/2023] Open
Abstract
Colorectal cancer liver metastasis (CRLM) is one of the leading causes of death among patients with colorectal cancer (CRC). Although immunotherapy has demonstrated encouraging outcomes in CRC, its benefits are minimal in CRLM. The complex immune landscape of the hepatic tumour microenvironment is essential for the development of a premetastatic niche and for the colonisation and metastasis of CRC cells; thus, an in-depth understanding of these mechanisms can provide effective immunotherapeutic targets for CRLM. This review summarises recent studies on the immune landscape of the tumour microenvironment of CRLM and highlights therapeutic prospects for targeting the suppressive immune microenvironment of CRLM.
Collapse
Affiliation(s)
- Yaxian Wang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Xinyang Zhong
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Xuefeng He
- ZJU-UCLA Joint Center for Medical Education and Research, Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zijuan Hu
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China
- Institute of Pathology, Fudan University, Shanghai, China
| | - Huixia Huang
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China
- Institute of Pathology, Fudan University, Shanghai, China
| | - Jiayu Chen
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Keji Chen
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China
- Institute of Pathology, Fudan University, Shanghai, China
| | - Senlin Zhao
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Ping Wei
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China.
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China.
- Institute of Pathology, Fudan University, Shanghai, China.
| | - Dawei Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China.
| |
Collapse
|
447
|
Sbrana A, Mazzini G, Comolli G, Antonuzzo A, Danova M. The contribution of automated cytometry in immuno-oncology. Methods Cell Biol 2023; 195:23-37. [PMID: 40180453 DOI: 10.1016/bs.mcb.2023.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
Cancer immunotherapy has been a real revolution and has given many survival chances to several patients. However, the understanding of resistance to immunotherapy is still an unmet need in clinical practice. Monitoring of immune mechanisms could be a tool to better understand this phenomenon. FCM and CyTOF could be used in this field, since they allow the simultaneous analysis of several protein expressions pattern, thus possibly understanding the functions of several immune cell populations, such as T cells, and their interactions with tumor cells and tumor microenvironment. Furthermore, automated cytometry could be used to understand the interaction of drugs with their target through the analysis of receptor occupancy. Spectral overlap, however, could be a limit for multiple simultaneous analyses. Other possible limitations of these techniques are a low number of cells in samples and the need for viable cells (with the possible interference of cell debris). The lack of standardized protocols, and thus the difficult reproducibility, have been the major limit to their application in clinical practice, so international efforts have been made to get to shared guidelines. Ongoing trials are to answer to the possibility of clinical application of these techniques.
Collapse
Affiliation(s)
- Andrea Sbrana
- Department of Surgical, Medical and Molecular Pathology and Critical Care Area, University of Pisa, Pisa, Italy; Service of Pneumo-Oncology, Unit of Pneumology, Pisa, Italy
| | | | - Giuditta Comolli
- Department of Microbiology and Virology and Laboratory of Biochemistry-Biotechnology and Advanced Diagnostics, IRCCS San Matteo Foundation, Pavia, Italy
| | | | - Marco Danova
- Unit of Internal Medicine and Medical Oncology, Vigevano Civic Hospital, Pavia, Italy; LIUC University, Castellanza, Varese, Italy.
| |
Collapse
|
448
|
Lucci A, Addanki S, Chiang YJ, Meas S, Sarli VN, Upshaw JR, Manchem M, Patel SP, Wargo JA, Gershenwald JE, Ross MI. Presence of Circulating Tumor Cells Predates Imaging Detection of Relapse in Patients with Stage III Melanoma. Cancers (Basel) 2023; 15:3630. [PMID: 37509290 PMCID: PMC10377914 DOI: 10.3390/cancers15143630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/07/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Stage III melanoma includes nodal metastasis or in-transit disease. Five-year survival rates vary between 32% and 93%. The identification of high-risk patients is important for clinical decision making. We demonstrated previously that ≥1 circulating tumor cells (CTCs) at baseline was associated with recurrence. In this study, we investigated how frequently CTCs were identified prior to radiologically detected recurrence. Stage III patients (n = 325) had imaging at baseline and q 3 months. Baseline and q 6-12 months blood draws (7.5 mL) were performed to identify CTCs up to 3.5 years from diagnosis. CTC assessment was performed using the immunomagnetic capture of CD146-positive cells and anti-MEL-PE. The presence of one or more CTCs was considered positive. We analyzed the cohort of patients with relapse confirmed by radiologic imaging. CTC collection dates were assessed to determine the lead time for CTC detection. CTC-negative patients were significantly less likely to relapse compared to patients positive for CTCs (p-value < 0.001). Within the 325-patient cohort, 143 patients (44%) had recurrence, with a median follow-up of 52 months from diagnosis. The cohort (n = 143) with positive imaging and CTC results revealed 76% of patients (108/143) had CTC+ results before the radiological identification of relapse. The median time between positive CTC and positive imaging was 9 months. CTCs were positive in >75% of patients prior to relapse at a median of 9 months before radiologic detection.
Collapse
Affiliation(s)
- Anthony Lucci
- Departments of Breast Surgical Oncology and Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sridevi Addanki
- Departments of Breast Surgical Oncology and Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yi-Ju Chiang
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Salyna Meas
- Departments of Breast Surgical Oncology and Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Vanessa N Sarli
- Departments of Breast Surgical Oncology and Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Joshua R Upshaw
- Departments of Breast Surgical Oncology and Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mayank Manchem
- Departments of Breast Surgical Oncology and Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sapna P Patel
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jennifer A Wargo
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jeffrey E Gershenwald
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Merrick I Ross
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
449
|
Ke G, Cheng N, Sun H, Meng X, Xu L. Explore the impact of hypoxia-related genes (HRGs) in Cutaneous melanoma. BMC Med Genomics 2023; 16:160. [PMID: 37422626 PMCID: PMC10329328 DOI: 10.1186/s12920-023-01587-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 06/20/2023] [Indexed: 07/10/2023] Open
Abstract
BACKGROUND Cutaneous melanoma (CM) has an overall poor prognosis due to a high rate of metastasis. This study aimed to explore the role of hypoxia-related genes (HRGs) in CM. METHODS We first used on-negative matrix factorization consensus clustering (NMF) to cluster CM samples and preliminarily analyzed the relationship of HRGs to CM prognosis and immune cell infiltration. Subsequently, we identified prognostic-related hub genes by univariate COX regression analysis and the least absolute shrinkage and selection operator (LASSO) and constructed a prognostic model. Finally, we calculated a risk score for patients with CM and investigated the relationship between the risk score and potential surrogate markers of response to immune checkpoint inhibitors (ICIs), such as TMB, IPS values, and TIDE scores. RESULTS Through NMF clustering, we identified high expression of HRGs as a risk factor for the prognosis of CM patients, and at the same time, increased expression of HRGs also indicated a poorer immune microenvironment. Subsequently, we identified eight gene signatures (FBP1, NDRG1, GPI, IER3, B4GALNT2, BGN, PKP1, and EDN2) by LASSO regression analysis and constructed a prognostic model. CONCLUSION Our study identifies the prognostic significance of hypoxia-related genes in melanoma and shows a novel eight-gene signature to predict the potential efficacy of ICIs.
Collapse
Affiliation(s)
- Guolin Ke
- Department of Dermatology and Venereology, Yijishan Hospital, Wannan Medical College, No. 2 Zheshan West Road, Wuhu City, Anhui Province, China
| | - Nan Cheng
- Department of Dermatology and Venereology, Yijishan Hospital, Wannan Medical College, No. 2 Zheshan West Road, Wuhu City, Anhui Province, China
| | - Huiya Sun
- Department of Dermatology and Venereology, Yijishan Hospital, Wannan Medical College, No. 2 Zheshan West Road, Wuhu City, Anhui Province, China
| | - Xiumei Meng
- Department of Dermatology and Venereology, Yijishan Hospital, Wannan Medical College, No. 2 Zheshan West Road, Wuhu City, Anhui Province, China
| | - Lei Xu
- Department of Hand, Foot, and Ankle Surgery, Yijishan Hospital, Wannan Medical College, No. 2 Zheshan West Road, Wuhu City, Anhui Province, China.
| |
Collapse
|
450
|
Dagar G, Gupta A, Masoodi T, Nisar S, Merhi M, Hashem S, Chauhan R, Dagar M, Mirza S, Bagga P, Kumar R, Akil ASAS, Macha MA, Haris M, Uddin S, Singh M, Bhat AA. Harnessing the potential of CAR-T cell therapy: progress, challenges, and future directions in hematological and solid tumor treatments. J Transl Med 2023; 21:449. [PMID: 37420216 PMCID: PMC10327392 DOI: 10.1186/s12967-023-04292-3] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 06/21/2023] [Indexed: 07/09/2023] Open
Abstract
Traditional cancer treatments use nonspecific drugs and monoclonal antibodies to target tumor cells. Chimeric antigen receptor (CAR)-T cell therapy, however, leverages the immune system's T-cells to recognize and attack tumor cells. T-cells are isolated from patients and modified to target tumor-associated antigens. CAR-T therapy has achieved FDA approval for treating blood cancers like B-cell acute lymphoblastic leukemia, large B-cell lymphoma, and multiple myeloma by targeting CD-19 and B-cell maturation antigens. Bi-specific chimeric antigen receptors may contribute to mitigating tumor antigen escape, but their efficacy could be limited in cases where certain tumor cells do not express the targeted antigens. Despite success in blood cancers, CAR-T technology faces challenges in solid tumors, including lack of reliable tumor-associated antigens, hypoxic cores, immunosuppressive tumor environments, enhanced reactive oxygen species, and decreased T-cell infiltration. To overcome these challenges, current research aims to identify reliable tumor-associated antigens and develop cost-effective, tumor microenvironment-specific CAR-T cells. This review covers the evolution of CAR-T therapy against various tumors, including hematological and solid tumors, highlights challenges faced by CAR-T cell therapy, and suggests strategies to overcome these obstacles, such as utilizing single-cell RNA sequencing and artificial intelligence to optimize clinical-grade CAR-T cells.
Collapse
Affiliation(s)
- Gunjan Dagar
- Department of Medical Oncology (Lab.), Dr. BRAIRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, Delhi, 110029, India
| | - Ashna Gupta
- Department of Medical Oncology (Lab.), Dr. BRAIRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, Delhi, 110029, India
| | - Tariq Masoodi
- Laboratory of Cancer Immunology and Genetics, Sidra Medicine, Doha, Qatar
| | - Sabah Nisar
- Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Maysaloun Merhi
- National Center for Cancer Care and Research, Hamad Medical Corporation, 3050, Doha, Qatar
| | - Sheema Hashem
- Department of Human Genetics, Sidra Medicine, Doha, Qatar
| | - Ravi Chauhan
- Department of Medical Oncology (Lab.), Dr. BRAIRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, Delhi, 110029, India
| | - Manisha Dagar
- Shiley Eye Institute, University of California San Diego, San Diego, CA, USA
| | - Sameer Mirza
- Department of Chemistry, College of Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Puneet Bagga
- Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Rakesh Kumar
- School of Biotechnology, Shri Mata Vaishno Devi University, Katra, Jammu and Kashmir, 182320, India
| | - Ammira S Al-Shabeeb Akil
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, P.O. Box 26999, Doha, Qatar
| | - Muzafar A Macha
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology, Pulwama, Jammu and Kashmir, India
| | - Mohammad Haris
- Center for Advanced Metabolic Imaging in Precision Medicine, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
- Laboratory Animal Research Center, Qatar University, Doha, Qatar
| | - Shahab Uddin
- Laboratory Animal Research Center, Qatar University, Doha, Qatar.
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, P.O. Box 3050, Doha, Qatar.
| | - Mayank Singh
- Department of Medical Oncology (Lab.), Dr. BRAIRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, Delhi, 110029, India.
| | - Ajaz A Bhat
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, P.O. Box 26999, Doha, Qatar.
| |
Collapse
|