1
|
Zhao L, Cheng X, Zhao H, Zhao H, Di W, Mei Z. Long‑term survival after comprehensive treatment in a patient with advanced neuroendocrine neoplasm of the pancreas: A case report. Oncol Lett 2025; 29:49. [PMID: 39564371 PMCID: PMC11574579 DOI: 10.3892/ol.2024.14795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 10/11/2024] [Indexed: 11/21/2024] Open
Abstract
Neuroendocrine neoplasms of the pancreas (pNENs) are rare. In February 2021, a 54-year-old woman was diagnosed with pNEN and multiple metastases within the liver. The patient, diagnosed with grade G2 neuroendocrine neoplasm (T4N0M1), underwent an ultrasonography-guided liver biopsy and radiofrequency ablation. After receiving Sandostatin LAR in April 2021, side effects led to its discontinuation after seven cycles. Following two sessions of radiofrequency ablation, the patient's condition was stable. However, disease progression was noted in September 2023, resulting in hemodialysis and closed peritoneal drainage. Surufatinib was administered, stabilizing the tumor by November 2023. The patient underwent transarterial chemoembolization due to a large tumor burden, with subsequent MRCP showing stability from diagnosis in February 2021 to June 2024. The present case report highlights the role of tailored treatment strategies considering patient comorbidities and tumor biology, and the significance of secondary puncture biopsy, which, despite not being pursued by the patient in the present study due to the associated risks, may provide survival benefits for patients with advanced or metastatic pNEN.
Collapse
Affiliation(s)
- Lei Zhao
- Department of Clinical Laboratory, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Xin Cheng
- Department of Clinical Laboratory, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Hongbin Zhao
- Department of General Surgery, Huaihai Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi 046000, P.R. China
| | - Haifei Zhao
- Department of Imaging, Huaihai Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi 046000, P.R. China
| | - Wenyu Di
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, P.R. China
| | - Zhihong Mei
- Department of Radiotherapy, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, P.R. China
| |
Collapse
|
2
|
Sasaki Y, Maeda T, Hojo M, Miura T, Ishikawa K, Funayama E, Okada K, Yamamoto Y. Synergistic anti-tumor effects of oncolytic virus and anti-programmed cell death protein 1 antibody combination therapy: For suppression of lymph node and distant metastasis in a murine melanoma model. Biochem Biophys Res Commun 2024; 740:151011. [PMID: 39571230 DOI: 10.1016/j.bbrc.2024.151011] [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: 08/20/2024] [Revised: 11/10/2024] [Accepted: 11/14/2024] [Indexed: 12/01/2024]
Abstract
It is believed that oncolytic viruses (OVs) exert both direct anti-tumor effects by intratumoral injection as well as indirect anti-tumor effects by activating systemic immunity. In phase III clinical trials, OV and anti-programmed cell death-1 (aPD-1) antibody combination therapy showed no significant differences in overall survival and progression-free survival in patients with unresectable advanced melanoma. In the study, OVs can exert only indirect anti-tumor effects in non-injected, systemic lesions. If the tumor is at a stage where both direct and indirect anti-tumor effects of OVs can be expected, OVs may further enhance the therapeutic effect, in addition to the clinically expected therapeutic effect. Therefore, we investigated whether canerpaturev (C-REV) and aPD-1 antibody combination therapy suppresses tumor progression in a murine melanoma model. Our findings showed that the C-REV and aPD-1 antibody combination therapy suppressed tumor progression in a murine melanoma model. The combination therapy stimulated systemic immunity in lymphoid tissues by activating helper T cells and B cells to enhance adaptive and humoral immunity, as well as by increasing effector/memory T cell fractions. Synergistically enhanced systemic anti-tumor effects suppressed lymph node and lung metastases. These findings suggest that direct anti-tumor effects by infecting and destroying cancer cells from within and indirect anti-tumor effects enhanced by the combination therapy worked simultaneously to suppress tumor progression. Our results may provide evidence to support the usefulness of OV and aPD-1 antibody combination therapy as a neoadjuvant therapy in the surgical treatment of melanoma.
Collapse
Affiliation(s)
- Yuki Sasaki
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Japan.
| | - Taku Maeda
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Japan.
| | - Masahiro Hojo
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Japan.
| | - Takahiro Miura
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Japan.
| | - Kosuke Ishikawa
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Japan.
| | - Emi Funayama
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Japan.
| | - Kazufumi Okada
- Data Science Center, Promotion Unit, Institute of Health Science Innovation for Medical Care, Hokkaido University Hospital, Japan.
| | - Yuhei Yamamoto
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Japan.
| |
Collapse
|
3
|
Acar C, Yüksel HÇ, Şahin G, Açar FP, Tünbekici S, Çelebi G, Karaca B. Efficacy and prognostic factors of anti-PD1 and nivolumab-ipilimumab therapy in advanced melanoma patients resistant to prior ICI treatment. Discov Oncol 2024; 15:813. [PMID: 39704850 DOI: 10.1007/s12672-024-01702-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 12/12/2024] [Indexed: 12/21/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) have significantly improved the five-year survival rate for advanced melanoma. However, many patients exhibit resistance to ICI therapy. This study evaluated the efficacy and prognostic factors of anti-PD-1 (Group A) and nivolumab-ipilimumab (Group B) therapy in patients with advanced melanoma who were resistant to prior ICI therapy. We conducted a retrospective analysis of 56 patients with advanced melanoma who had previously shown resistance to ICI therapy. In the Group A (who have previously shown resistance to anti-CTLA-4, n = 28), the objective response rate (ORR) was 42.9%, with a disease control rate (DCR) of 53%. In the Group B (previously shown resistance to anti-PD-1, n = 28), the ORR was 17.9%, and the DCR was 25%. The ORR was lower in two subgroups: patients who showed progression or relapse in the the initial radiological assessment of prior ICI therapy (ORR 10.5%) and patients who had previously received ICI in the adjuvant setting (ORR 8.3%). A Royal Marsden Hospital (RMH) score of 2-3 was a predictor of OS in both groups (Group A: HR 3.789, 95% CI 1.356-10.589, p = 0.011; Group B: HR 4.281, 95% CI 1.490-12.300, p = 0.007) and for PFS in the Group B (HR 3.167, 95% CI 1.062-9.442, p = 0.039). Anti-PD-1 therapy demonstrated efficacy following resistance to anti-CTLA-4, whereas combination ICI therapy showed lower response rates in patients resistant to anti-PD-1. Further studies are needed to confirm the RMH scores and other prognostic markers and to evaluate subgroups with lower efficacy of nivolumab-ipilimumab therapy.
Collapse
Affiliation(s)
- Caner Acar
- Division of Medical Oncology, Department of Internal Medicine, Ege University Medical Faculty, Izmir, 35100, Turkey.
| | - Haydar Çağatay Yüksel
- Division of Medical Oncology, Department of Internal Medicine, Ege University Medical Faculty, Izmir, 35100, Turkey
| | - Gökhan Şahin
- Division of Medical Oncology, Department of Internal Medicine, Ege University Medical Faculty, Izmir, 35100, Turkey
| | - Fatma Pinar Açar
- Division of Medical Oncology, Department of Internal Medicine, Ege University Medical Faculty, Izmir, 35100, Turkey
| | - Salih Tünbekici
- Division of Medical Oncology, Department of Internal Medicine, Ege University Medical Faculty, Izmir, 35100, Turkey
| | - Gülçin Çelebi
- Department of Internal Medicine, Ege University Medical Faculty, Izmir, Turkey, 35100
| | - Burçak Karaca
- Division of Medical Oncology, Department of Internal Medicine, Ege University Medical Faculty, Izmir, 35100, Turkey
| |
Collapse
|
4
|
Tawbi HA, Hodi FS, Lipson EJ, Schadendorf D, Ascierto PA, Matamala L, Castillo Gutiérrez E, Rutkowski P, Gogas H, Lao CD, Janoski De Menezes J, Dalle S, Arance AM, Grob JJ, Ratto B, Rodriguez S, Mazzei A, Dolfi S, Long GV. Three-Year Overall Survival With Nivolumab Plus Relatlimab in Advanced Melanoma From RELATIVITY-047. J Clin Oncol 2024:JCO2401124. [PMID: 39671533 DOI: 10.1200/jco.24.01124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 09/23/2024] [Accepted: 10/14/2024] [Indexed: 12/15/2024] Open
Abstract
Nivolumab plus relatlimab demonstrated a statistically significant improvement in progression-free survival (PFS), along with a clinically meaningful, but not statistically significant improvement in overall survival (OS) and a numerically higher objective response rate (ORR) compared with nivolumab in the RELATIVITY-047 trial (ClinicalTrials.gov identifier: NCT03470922). We report updated descriptive efficacy and safety results from RELATIVITY-047 with a median follow-up of 33.8 months. Median PFS was 10.2 months (95% CI, 6.5 to 15.4) with nivolumab plus relatlimab and 4.6 months (95% CI, 3.5 to 6.5) with nivolumab (hazard ratio [HR], 0.79 [95% CI, 0.66 to 0.95]); median OS was 51.0 months (95% CI, 34.0 to not reached) and 34.1 (95% CI, 25.2 to 44.7) months, respectively (HR, 0.80 [95% CI, 0.66 to 0.99]). ORR was 43.7% (95% CI, 38.4 to 49.0) with nivolumab plus relatlimab and 33.7% (95% CI, 28.8 to 38.9) with nivolumab. Efficacy across the majority of prespecified subgroups favored the combination. No new or unexpected safety signals were identified. Overall, at 3-year follow-up, the benefit observed with nivolumab plus relatlimab compared with nivolumab in patients with advanced melanoma was sustained, with the OS HR 95% CI upper bound now <1. This benefit is accompanied by a safety profile consistent with previous reports.
Collapse
Affiliation(s)
- Hussein A Tawbi
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Evan J Lipson
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins Medicine, The Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Dirk Schadendorf
- University of Essen and the German Cancer Consortium, Essen, Germany
| | - Paolo A Ascierto
- Istituto Nazionale dei Tumori IRCCS "Fondazione G. Pascale," Naples, Italy
| | - Luis Matamala
- Instituto Oncológico Fundación Arturo López Pérez and Department of Oncology, Instituto Nacional dfoel Cáncer, Santiago, Chile
| | | | - Piotr Rutkowski
- Maria Skłodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Helen Gogas
- National and Kapodistrian University of Athens, Athens, Greece
| | - Christopher D Lao
- Michigan Medicine, Rogel Cancer Center, University of Michigan, Ann Arbor, MI
| | | | - Stéphane Dalle
- Hospices Civils de Lyon, Cancer Research Center of Lyon, Pierre-Bénite, France
| | | | | | | | | | | | | | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, Royal North Shore and Mater Hospitals, Sydney, NSW, Australia
| |
Collapse
|
5
|
Wang L, Hu Z, Zhang W, Wang Z, Cao M, Cao X. Promoting macrophage phagocytosis of cancer cells for effective cancer immunotherapy. Biochem Pharmacol 2024; 232:116712. [PMID: 39675588 DOI: 10.1016/j.bcp.2024.116712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 11/26/2024] [Accepted: 12/10/2024] [Indexed: 12/17/2024]
Abstract
Cancer therapy has been revolutionized by immunotherapeutic agents exploiting adaptive antitumor immunity in the past two decades. However, the overall response rate of these immunotherapies is limited, and patients also develop resistance upon treatment, promoting a rapidly growing exploration of anti-tumor innate immunity for effective cancer therapy. Among these, macrophage immunotherapy through harnessing macrophage phagocytosis has been thrust into the spotlight due to its potential for simultaneously inducing cancer cell killing effect and mobilizing adaptive antitumor responses. Here in this review, we summarize the current macrophage immunotherapy such as therapeutic antibodies, phagocytosis checkpoint blockades, and CAR-macrophages with a particular emphasis on the resistant mechanisms limiting their therapeutic effects. Moreover, we further survey the efforts being placed to seek synergistic mechanisms and combination strategies for promoting macrophage phagocytosis which might stand as next-generation cancer immunotherapy.
Collapse
Affiliation(s)
- Lei Wang
- Shanghai Frontiers Science Center for Drug Target Identification and Delivery, and the Engineering Research Center of Cell and Therapeutic Antibody of the Ministry of Education, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai, China
| | - Ziyi Hu
- Shanghai Frontiers Science Center for Drug Target Identification and Delivery, and the Engineering Research Center of Cell and Therapeutic Antibody of the Ministry of Education, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai, China
| | - Wencan Zhang
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Zhixin Wang
- Shanghai Frontiers Science Center for Drug Target Identification and Delivery, and the Engineering Research Center of Cell and Therapeutic Antibody of the Ministry of Education, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai, China
| | - Ming Cao
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xu Cao
- Shanghai Frontiers Science Center for Drug Target Identification and Delivery, and the Engineering Research Center of Cell and Therapeutic Antibody of the Ministry of Education, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
6
|
Azimnasab-Sorkhabi P, Soltani-Asl M, Bouhajra M, Ansa-Addo EA, Junior JRK. Soluble CTLA-4 - A confounding factor in CTLA-4 based checkpoint immunotherapy in cancer. Immunol Lett 2024:106965. [PMID: 39667581 DOI: 10.1016/j.imlet.2024.106965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 11/30/2024] [Accepted: 12/09/2024] [Indexed: 12/14/2024]
Abstract
Cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) is a receptor that inhibits the activity of T cells. The CTLA-4 gene consists of four different exons that enable four different isoforms of CTLA-4 to be generated through alternative splicing. Although sCTLA-4 might impede the therapeutic effect of anti-CTLA-4 treatments, the role of sCTLA-4 in the tumor microenvironment (TME) is not well understood. Here, we provide novel perspectives on the inhibitory characteristics of sCTLA-4 in TME.
Collapse
Affiliation(s)
- Parviz Azimnasab-Sorkhabi
- Department of Surgery, School of Veterinary Medicine and Animal Sciences, University of Sao Paulo, Sao Paulo, Brazil; Pelotonia Institute for Immuno-Oncology and Division of Medical Oncology, Department of Internal Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, USA.
| | - Maryam Soltani-Asl
- Department of Surgery, School of Veterinary Medicine and Animal Sciences, University of Sao Paulo, Sao Paulo, Brazil; Pelotonia Institute for Immuno-Oncology and Division of Medical Oncology, Department of Internal Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Musab Bouhajra
- Pelotonia Institute for Immuno-Oncology and Division of Medical Oncology, Department of Internal Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Ephraim A Ansa-Addo
- Pelotonia Institute for Immuno-Oncology and Division of Medical Oncology, Department of Internal Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Jose Roberto Kfoury Junior
- Department of Surgery, School of Veterinary Medicine and Animal Sciences, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
7
|
Fu C, Gu H, Sun L, Wang Z, Zhang Q, Luo N, Chen D, Zhou T. Predictive value of ZFHX4 mutation for the efficacy of immune checkpoint inhibitors in non-small cell lung cancer and melanoma. Invest New Drugs 2024; 42:623-634. [PMID: 39369144 DOI: 10.1007/s10637-024-01477-5] [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: 07/10/2024] [Accepted: 09/26/2024] [Indexed: 10/07/2024]
Abstract
Studies have shown that the Zinc finger homeobox 4 (ZFHX4) might be a factor in the prognosis of malignancies. However, little is known about the association between the ZFHX4 mutation and the effectiveness of immune checkpoint inhibitors (ICIs) in non-small cell lung cancer (NSCLC) and melanoma. Three public ICIs-treated NSCLC cohorts were divided into discovery cohort (n=75) and validation cohort (n=62), which were used to evaluate the relationship between ZFHX4 mutation and ICIs effectiveness in NSCLC. Seven ICIs-treated melanoma cohorts (n = 418) were used to analyze the relationship between ZFHX4 mutation and immunotherapy efficacy in melanoma. NSCLC and skin cutaneous melanoma (SKCM) cohorts from The Cancer Genome Atlas (TCGA) were used to investigate underlying mechanism. Patients with ZFHX4 mutant-type (ZFHX4-Mut) showed a superior objective response rate (ORR) (P < 0.01) and longer progression-free survival (PFS) (P < 0.05) than patients with ZFHX4 wild-type (ZFHX4-WT) in NSCLC cohorts. In the melanoma cohorts, patients carrying ZFHX4-Mut had a higher ORR (P = 0.042) and longer overall survival (OS) (P = 0.011). Besides, patients with NSCLC and melanoma harboring ZFHX4-Mut had a higher tumor mutation burden (TMB) (P<0.001) and tumor neoantigen burden (TNB) (P<0.001) than those harboring ZFHX4-WT. ZFHX4 mutation was associated with higher levels of plasma B cells, activated CD4+ memory T cells, and CD8+ T cells. Seven DNA damage repair pathways were significantly enriched in the ZFHX4-Mut group. ZFHX4 mutation could serve as a predicter for the efficacy of ICIs therapy in NSCLC and melanoma.
Collapse
Affiliation(s)
- Cong Fu
- Department of Oncology, Changzhou Cancer (Fourth People's) Hospital, Changzhou, 213000, China
| | - Haoran Gu
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, 221004, China
| | - Lin Sun
- Department of Oncology, Changzhou Cancer (Fourth People's) Hospital, Changzhou, 213000, China
| | - Zhouyu Wang
- The State Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd., Nanjing, 210002, China
| | - Qin Zhang
- The State Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd., Nanjing, 210002, China
| | - Ningning Luo
- The State Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd., Nanjing, 210002, China
| | - Dongsheng Chen
- The State Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd., Nanjing, 210002, China.
- Cancer Center, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, China.
- Center of Translational Medicine, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, China.
| | - Tong Zhou
- Department of Oncology, Changzhou Cancer (Fourth People's) Hospital, Changzhou, 213000, China.
| |
Collapse
|
8
|
Wang S, Liu C, Yang C, Jin Y, Cui Q, Wang D, Ge T, He G, Li W, Zhang G, Liu A, Xia Y, Liu Y, Yu J. PI3K/AKT/mTOR and PD‑1/CTLA‑4/CD28 pathways as key targets of cancer immunotherapy (Review). Oncol Lett 2024; 28:567. [PMID: 39390982 PMCID: PMC11465225 DOI: 10.3892/ol.2024.14700] [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: 06/21/2024] [Accepted: 08/08/2024] [Indexed: 10/12/2024] Open
Abstract
T cells play an important role in cancer, and energy metabolism can determine both the proliferation and differentiation of T cells. The inhibition of immune checkpoint molecules programmed cell death protein 1 (PD-1) and cytotoxic T-lymphocyte associated protein 4 (CTLA-4) are a promising cancer treatment. In recent years, research on CD28 has increased. Although numerous reports involve CD28 and its downstream PI3K/AKT/mTOR signaling mechanisms in T cell metabolism, they have not yet been elucidated. A literature search strategy was used for the databases PubMed, Scopus, Web of Science and Cochrane Library to ensure broad coverage of medical and scientific literature, using a combination of keywords including, but not limited to, 'lung cancer' and 'immunotherapy'. Therefore, the present study reviewed the interaction and clinical application of the PD-1/CTLA-4/CD28 and PI3K/AKT/mTOR pathways in T cells, aiming to provide a theoretical basis for immunotherapy in clinical cancer patients.
Collapse
Affiliation(s)
- Shuangcui Wang
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 301617, P.R. China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Changyu Liu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
- School of Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Chenxin Yang
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Yutong Jin
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Qian Cui
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 301617, P.R. China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Dong Wang
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 301617, P.R. China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Ting Ge
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 301617, P.R. China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Guixin He
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 301617, P.R. China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Wentao Li
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 301617, P.R. China
| | - Guan Zhang
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 301617, P.R. China
| | - Aqing Liu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 301617, P.R. China
| | - Ying Xia
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 301617, P.R. China
| | - Yunhe Liu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 301617, P.R. China
| | - Jianchun Yu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 301617, P.R. China
| |
Collapse
|
9
|
Zhang X, Deng J, Wu R, Hu J. Manganese improves anti-PD-L1 immunotherapy via eliciting type I interferon signaling in melanoma. Invest New Drugs 2024; 42:685-693. [PMID: 39592531 DOI: 10.1007/s10637-024-01484-6] [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: 11/18/2024] [Indexed: 11/28/2024]
Abstract
The immune checkpoint inhibitor therapy represented by blocking programmed cell death protein 1/ programmed cell death-ligand 1 (PD-1/PD-L1) has made significant progress in melanoma treatment. However, the response rate and therapeutic effect of immunotherapy alone are still not ideal for melanoma. In this study, we aimed to evaluate the defects of treating anti-PD-L1 alone and the therapeutic effect and molecular mechanism of combined therapy with anti-PD-L1 and MnCl2. We detected the changes of immune cell populations after anti-PD-L1 treatment in melanoma xenograft mouse model. Further, we evaluated the regulatory effect of MnCl2 on dendritic cells (DCs) maturation in vitro. Next, we tested the therapeutic effect and regulatory effect on the tumor microenvironment with anti-PD-L1 and MnCl2 via combining treatment with anti-PD-L1 and MnCl2. Anti-PD-L1 therapy has a certain tumor suppressive function, but the effect is not ideal. The results of flow cytometry showed that the number of CD4+ T cells and CD8+ T cells significantly increased after anti-PD-L1 treatment. However, the number of DCs remained basically unchanged after anti-PD-L1 treatment. In vitro, we confirmed that MnCl2 significantly promoted DCs maturation vis activating cGAS-STING signaling pathway. The combination of anti-PD-L1 and MnCl2 displayed the best tumor suppression effect in melanoma xenograft mouse model. In tumor microenvironment, the infiltration of T cells and the maturation of DCs were significantly promoted, demonstrating a strong anti-tumor immune response. In summary, we conclude that combining anti-PD-L1 with MnCl2 is a promising therapeutic strategy for melanoma.
Collapse
Affiliation(s)
- Xiaoxin Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jianhua Deng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Renjie Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jian Hu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
| |
Collapse
|
10
|
Dasanu CA, Plaxe SC. Temozolomide overcoming resistance to immune checkpoint inhibitors in relapsed/refractory metastatic melanoma? Insights from a single center series. J Oncol Pharm Pract 2024:10781552241302421. [PMID: 39584741 DOI: 10.1177/10781552241302421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2024]
Abstract
There is a need to develop more effective salvage therapies for patients with relapsed melanoma of the skin. Research has shown that chemotherapy-induced cancer cell death may increase immunogenic antigen exposure, or upregulation of co-inhibitory ligands such as PD-L1, thereby augmenting immune checkpoint inhibitor (ICI) efficacy. In addition, chemotherapy preconditioning may lead to depletion of Tregs, known to suppress immune anti-melanoma responses. As a result, regimens including both chemotherapy and ICI constructs are currently successfully employed in the 1st line therapy of many solid tumors. We report a series of three patients with metastatic melanoma, refractory to ICI treatment, who responded to salvage therapy with temozolomide (TMZ) in combination with PD-1 inhibitors, with or without CTLA-4 inhibitors. The responses were durable, each lasting more than 12 months. In two patients, complete responses are ongoing at 13 and 15 months, respectively. Randomized clinical trials with TMZ plus ICIs for patients with relapsed or refractory malignant melanoma seem warranted.
Collapse
Affiliation(s)
- Constantin A Dasanu
- Lucy Curci Cancer Center, Eisenhower Health, Rancho Mirage, CA, USA
- Department of Medical Oncology, UC San Diego Health System, San Diego, CA, USA
| | - Steven C Plaxe
- Lucy Curci Cancer Center, Eisenhower Health, Rancho Mirage, CA, USA
- Department of Medical Oncology, UC San Diego Health System, San Diego, CA, USA
| |
Collapse
|
11
|
Cao Y, Zhao X, Miao Y, Wang X, Deng D. How the Versatile Self-Assembly in Drug Delivery System to Afford Multimodal Cancer Therapy? Adv Healthc Mater 2024:e2403715. [PMID: 39587000 DOI: 10.1002/adhm.202403715] [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: 09/27/2024] [Revised: 11/04/2024] [Indexed: 11/27/2024]
Abstract
The rapid development of self-assembly technology during the past few decades has effectively addressed plenty of the issues associated with carrier-based drug delivery systems, such as low loading efficiency, complex fabrication processes, and inherent toxicity of carriers. The integration of nanoscale delivery systems with self-assembly techniques has enabled efficient and targeted self-administration of drugs, enhanced bioavailability, prolonged circulation time, and controllable drug release. Concurrently, the limitations of single-mode cancer treatment, including low bioavailability, poor therapeutic outcomes, and significant side effects, have highlighted the urgent need for multimodal combined antitumor therapies. Set against the backdrop of multimodal cancer therapy, this review summarizes the research progress and applications of a large number of self-assembled drug delivery platforms, including natural small molecule self-assembled, carrier-free self-assembled, amphiphilic polymer-based self-assembled, peptide-based self-assembled, and metal-based self-assembled nano drug delivery systems. This review particularly analyzes the latest advances in the application of self-assembled nano drug delivery platforms in combined antitumor therapies mediated by chemotherapy, phototherapy, radiotherapy, sonodynamic therapy, and immunotherapy, providing innovative research insights for further optimization and expansion of self-assembled nano drug delivery systems in the clinical translation and development of antitumor combined therapy.
Collapse
Affiliation(s)
- Yuqi Cao
- Department of Pharmaceutical Engineering and Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Xiaomin Zhao
- Department of Pharmaceutical Engineering and Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Yuhang Miao
- Department of Pharmaceutical Engineering and Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Xin Wang
- Department of Pharmaceutical Engineering and Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Dawei Deng
- Department of Pharmaceutical Engineering and Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| |
Collapse
|
12
|
Margolin KA. Using Indirect Comparisons of Prospective, Randomized Trials to Make Therapeutic Decisions in Melanoma: Cross-Trial Comparisons as Surrogates for Proper Head-To-Head Studies? J Clin Oncol 2024; 42:3891-3894. [PMID: 39374466 DOI: 10.1200/jco-24-01634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 08/26/2024] [Accepted: 08/28/2024] [Indexed: 10/09/2024] Open
|
13
|
Amaral T, Ottaviano M, Arance A, Blank C, Chiarion-Sileni V, Donia M, Dummer R, Garbe C, Gershenwald JE, Gogas H, Guckenberger M, Haanen J, Hamid O, Hauschild A, Höller C, Lebbé C, Lee RJ, Long GV, Lorigan P, Muñoz Couselo E, Nathan P, Robert C, Romano E, Schadendorf D, Sondak V, Suijkerbuijk KPM, van Akkooi ACJ, Michelin O, Ascierto PA. Cutaneous melanoma: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol 2024:S0923-7534(24)04912-3. [PMID: 39550033 DOI: 10.1016/j.annonc.2024.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 11/07/2024] [Accepted: 11/08/2024] [Indexed: 11/18/2024] Open
Affiliation(s)
- T Amaral
- Skin Cancer Clinical Trials Center-University of Tuebingen, Tuebingen, Germany
| | - M Ottaviano
- Department of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione "G. Pascale", Napoli, Italy
| | - A Arance
- Department of Medical Oncology and IDIBAPS, Hospital Clinic y Provincial de Barcelona, Barcelona, Spain
| | - C Blank
- Department of Medical Oncology and Division of Immunology, The Netherlands Cancer Institute Antoni van Leeuwenhoek Ziekenhuis (NKI), Amsterdam; Leiden University Medical Center (LUMC), Leiden, The Netherlands; University Clinic Regensburg, Regensburg, Germany
| | - V Chiarion-Sileni
- Department of Oncology, Melanoma Unit, Istituto Oncologico Veneto, IOV-IRCCS, Padova, Italy
| | - M Donia
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital Herlev and Gentofte, Herlev, Denmark
| | - R Dummer
- Department of Dermatology, Skin Cancer Center, USZ-University Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - C Garbe
- Department of Dermatology, Center for DermatoOncology, University Hospital Tuebingen, Tuebingen, Germany
| | - J E Gershenwald
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center and The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, USA
| | - H Gogas
- First Department of Medicine, School of Medicine, National and Kapodistrian University of Athens-School of Medicine, Athens, Greece
| | - M Guckenberger
- Department of Radiation Oncology, University Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - J Haanen
- Division of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands; Department of Medical Oncology, Leiden University Medical Center (LUMC), Leiden, The Netherlands; Oncology Service, Melanoma Clinic, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - O Hamid
- Medical Oncology, Cutaneous Malignancies, The Angeles Clinic and Research Institute, A Cedars Sinai Affiliate, Los Angeles, USA
| | - A Hauschild
- Department of Dermatology, UKSH-Universitätsklinikum Schleswig-Holstein-Campus Kiel, Kiel, Germany
| | - C Höller
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - C Lebbé
- Université Paris Cite, AP-HP Dermato-oncology and CIC, Cancer Institute APHP, Nord Paris Cité, INSERM U976, Saint Louis Hospital, Paris, France
| | - R J Lee
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK; Faculty of Biology Medicine and Health, The University of Manchester, Manchester, UK
| | - G V Long
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia; Department Medical Oncology, Royal North Shore Hospital, Sydney, Australia; Mater Hospital, Sydney, Australia
| | - P Lorigan
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK; Faculty of Biology Medicine and Health, The University of Manchester, Manchester, UK
| | - E Muñoz Couselo
- Department of Medical Oncology, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - P Nathan
- Mount Vernon Cancer Centre, Northwood, UK
| | - C Robert
- Department of Oncology, Institut Gustave Roussy and Paris-Saclay University, Villejuif, France
| | - E Romano
- Department of Oncology, Center for Cancer Immunotherapy, Institut Curie, Paris, France
| | - D Schadendorf
- Department of Dermatology, WTZ-Westdeutsches Tumorzentrum Essen, National Center for Tumor Diseases (NCT-West), Campus Essen, Essen, Germany; University Alliance Ruhr, Research Center One Health, University Duisburg-Essen, Essen, Germany
| | - V Sondak
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, USA
| | - K P M Suijkerbuijk
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - A C J van Akkooi
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia; Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Sydney, Australia
| | - O Michelin
- Department of Oncology, Geneva University Hospital, Geneva, Switzerland
| | - P A Ascierto
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Instituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy
| |
Collapse
|
14
|
Márquez-Rodas I, Muñoz Couselo E, Rodríguez Moreno JF, Arance Fernández AM, Berciano Guerrero MÁ, Campos Balea B, de la Cruz Merino L, Espinosa Arranz E, García Castaño A, Berrocal Jaime A. SEOM-GEM clinical guidelines for cutaneous melanoma (2023). Clin Transl Oncol 2024; 26:2841-2855. [PMID: 38748192 PMCID: PMC11467041 DOI: 10.1007/s12094-024-03497-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 10/11/2024]
Abstract
Cutaneous melanoma incidence is rising. Early diagnosis and treatment administration are key for increasing the chances of survival. For patients with locoregional advanced melanoma that can be treated with complete resection, adjuvant-and more recently neoadjuvant-with targeted therapy-BRAF and MEK inhibitors-and immunotherapy-anti-PD-1-based therapies-offer opportunities to reduce the risk of relapse and distant metastases. For patients with advanced disease not amenable to radical treatment, these treatments offer an unprecedented increase in overall survival. A group of medical oncologists from the Spanish Society of Medical Oncology (SEOM) and Spanish Multidisciplinary Melanoma Group (GEM) has designed these guidelines, based on a thorough review of the best evidence available. The following guidelines try to cover all the aspects from the diagnosis-clinical, pathological, and molecular-staging, risk stratification, adjuvant therapy, advanced disease therapy, and survivor follow-up, including special situations, such as brain metastases, refractory disease, and treatment sequencing. We aim help clinicians in the decision-making process.
Collapse
Affiliation(s)
| | - Eva Muñoz Couselo
- Hospital Vall d'Hebron & Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | | | | | | | | | - Luis de la Cruz Merino
- Cancer Immunotherapy, Biomedicine Institute of Seville (IBIS)/CSIC, Clinical Oncology Department, University Hospital Virgen Macarena and School of Medicine, University of Seville, Seville, Spain
| | | | | | | |
Collapse
|
15
|
Ou Y, Liang S, Gao Q, Shang Y, Liang J, Zhang W, Liu S. Prognostic value of inflammatory markers NLR, PLR, LMR, dNLR, ANC in melanoma patients treated with immune checkpoint inhibitors: a meta-analysis and systematic review. Front Immunol 2024; 15:1482746. [PMID: 39493767 PMCID: PMC11527641 DOI: 10.3389/fimmu.2024.1482746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 09/30/2024] [Indexed: 11/05/2024] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) are an emerging tumor treatment pathway after traditional surgery, chemoradiotherapy, and targeted therapy. They have proven to be effective in a variety of cancers, but may not respond to non-target populations. Inflammatory markers such as neutrophil to lymphocyte ratio (NLR), platelet to lymphocyte ratio (PLR), lymphocyte to monocyte ratio (LMR), derived neutrophil lymphocyte ratio (dNLR), and neutrophil count (ANC) have been shown to be strongly associated with tumor prognosis, but their prognostic significance remains controversial. We therefore performed a meta-analysis to explore the association between NLR, PLR, LMR, dNLR, ANC and prognostic and clinicopathological factors in melanoma patients treated with ICIs. Methods A comprehensive search was conducted in Pubmed, Embase, Web Of Science and Cochrane databases, and the last search time was July 2024. To estimate the prognostic value of NLR, PLR, LMR, dNLR, ANC for PFS and OS, hazard ratio (HR) and corresponding 95% confidence interval (CI) estimates were used. Results This meta-analysis ultimately included 22 cohort studies involving 3235 melanoma patients. Meta-analysis results showed that high levels of NLR in melanoma patients receiving ICIs were associated with poorer OS and PFS, Merging the HR respectively OS [HR = 2.21, 95% CI (1.62, 3.02), P < 0.001], PFS [HR = 1.80, 95% CI (1.40, 2.30), P < 0.001]; High levels of PLR were associated with poor OS and PFS, and the combined HR was OS[HR=2.15,95%CI(1.66,2.80),P < 0.001] and PFS[HR=1.67,95%CI(1.31,2.12),P < 0.001]. High levels of dNLR were associated with poor OS and PFS, with combined HR being OS[HR=2.34,95%CI(1.96,2.79),P < 0.001] and PFS[HR=2.05,95%CI(1.73,2.42),P < 0.001], respectively. High ANC was associated with poor OS and PFS, and combined HR was OS[HR=1.95,95%CI(1.16,3.27),P < 0.001] and PFS[HR=1.63,95%CI(1.04,2.54),P=0.032], respectively. Increased LMR was associated with prolonged OS and PFS, with combined HR being OS[HR=0.36, 95%CI(0.19,0.70),P < 0.001] and PFS[HR=0.56,95%CI(0.40,0.79),P=0.034], respectively. Conclusion In melanoma patients treated with ICIs, elevated levels of NLR, PLR, dNLR, and ANC were associated with poorer overall survival OS and PFS. Conversely, a high LMR correlated with improved OS and PFS. Subgroup analyses indicated that dNLR may be linked to a worse prognosis in melanoma patients. In summary, inflammatory markers such as NLR, PLR, LMR, dNLR, and ANC serve as effective biomarkers for the prognostic assessment of melanoma patients following ICI treatment. These markers provide valuable insights for treatment decision-making in the realm of melanoma immunotherapy, and we anticipate further high-quality prospective studies to validate our findings in the future. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/#recordDetails, identifier CRD42024573406.
Collapse
Affiliation(s)
- Yan Ou
- Department of Plastic and Aesthetic Surgery, Affiliated Hospital of Shaanxi University of Chinese Medicine, Shaanxi, China
| | - Shufang Liang
- Department of Burns and Plastic Surgery, 969th Hospital of PLA Joint Logistic Support Force, Inner Mongolia, China
| | - Qiangqiang Gao
- Department of Proctology, Affiliated Hospital of Shaanxi University of Chinese Medicine, Shaanxi, China
| | - Yongran Shang
- The First Clinical Medical College of Shaanxi University of Chinese Medicine, Shaanxi, China
| | - Junfang Liang
- Department of Plastic and Aesthetic Surgery, Affiliated Hospital of Shaanxi University of Chinese Medicine, Shaanxi, China
| | - Weitao Zhang
- Department of Plastic and Aesthetic Surgery, Affiliated Hospital of Shaanxi University of Chinese Medicine, Shaanxi, China
| | - Sha Liu
- Department of Plastic and Aesthetic Surgery, Affiliated Hospital of Shaanxi University of Chinese Medicine, Shaanxi, China
| |
Collapse
|
16
|
Fujimura T, Yoshino K, Kato H, Fukushima S, Ishizuki S, Otsuka A, Matsushita S, Amagai R, Muto Y, Yamazaki E, Kambayashi Y, Yahata T, Miyata T, Fujisawa Y, Asano Y. A phase II multicentre study of plasminogen activator inhibitor-1 inhibitor (TM5614) plus nivolumab for treating anti-programmed cell death 1 antibody-refractory malignant melanoma: TM5614-MM trial. Br J Dermatol 2024; 191:691-697. [PMID: 38833158 DOI: 10.1093/bjd/ljae231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/12/2024] [Accepted: 05/25/2024] [Indexed: 06/06/2024]
Abstract
BACKGROUND Anti-programmed cell death 1 antibodies (PD-1 Abs) are widely used for advanced melanoma, but information on the efficacy of anti-PD-1 Abs is limited in the Asian population. There remains an unmet need to improve the therapeutic effects of anti-PD-1 Ab-treatment, particularly in patients with melanoma who are refractory to anti-PD-1 Abs. The aim of this study was to evaluate anti-PD-1 Ab-treatment in combination with TM5614 (a plasminogen activator inhibitor-1 inhibitor) in patients with unresectable melanoma. METHODS The TM5614-MM study was a multicentre, open-label, single-arm, phase II clinical trial to evaluate the efficacy and safety of nivolumab in combination with TM5614 in patients with advanced, unresectable malignant melanoma recruited at seven Japanese institutes between 13 September 2021 and 31 March 2023. Patients with metastatic or unresectable melanoma previously treated with anti-PD-1 Abs were enrolled. Nivolumab 480 mg was administered intravenously every 4 weeks for 8 weeks, while TM5614 was administered orally at a dose of 120 mg (0-4 weeks) and 180 mg once daily (5-8 weeks). The primary endpoint was the overall response rate after 8 weeks of concomitant use of TM5614. RESULTS Thirty-nine patients were enrolled, and 34 patients were included in the anti-PD-1 Ab-refractory cohort. The overall response rate at 8 weeks was 25.9% (95% confidence interval 12.9-44.9%, P = 0.027) in 27 patients who were anti-PD-1 Ab-refractory based on investigator assessment in the protocol per set cohort. Seven patients discontinued treatment owing to progressive disease or adverse events. Treatment-related grade 3 or higher adverse events occurred in 3 of 39 patients (7.7%) in the intention-to-treat cohort. CONCLUSIONS TM5614 in combination with nivolumab is well tolerated and effective in anti-PD-1 Ab-refractory unresectable melanoma.
Collapse
Affiliation(s)
- Taku Fujimura
- Department of Dermatology, Tohoku University School of Medicine, Sendai, Japan
| | - Koji Yoshino
- Department of Dermato-Oncology/Dermatology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo,Japan
- Department of Dermato-Oncology/Dermatology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Hiroshi Kato
- Department of Geriatric and Environmental Dermatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Satoshi Fukushima
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Shoichiro Ishizuki
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Atsushi Otsuka
- Department of Dermatology, Kindai University Hospital, Osaka, Japan
| | - Shigeto Matsushita
- Department of Dermato-Oncology/Dermatology, NHO Kagoshima Medical Center, Kagoshima, Japan
| | - Ryo Amagai
- Department of Dermatology, Tohoku University School of Medicine, Sendai, Japan
| | - Yusuke Muto
- Department of Dermatology, Tohoku University School of Medicine, Sendai, Japan
| | - Emi Yamazaki
- Department of Dermatology, Tohoku University School of Medicine, Sendai, Japan
| | - Yumi Kambayashi
- Department of Dermatology, Tohoku University School of Medicine, Sendai, Japan
| | - Takashi Yahata
- Translational Molecular Therapeutic Laboratory, Division of Host Defence Mechanism, Tokai University School of Medicine, Isehara,Japan
| | - Toshio Miyata
- Department of Molecular Medicine and Therapy, Tohoku University School of Medicine, Sendai, Japan
| | - Yasuhiro Fujisawa
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
- Department of Dermatology, Ehime University, Matsuyama, Japan
| | - Yoshihide Asano
- Department of Dermatology, Tohoku University School of Medicine, Sendai, Japan
| |
Collapse
|
17
|
Xu Y, Xiong Y. Targeting STING signaling for the optimal cancer immunotherapy. Front Immunol 2024; 15:1482738. [PMID: 39450170 PMCID: PMC11500076 DOI: 10.3389/fimmu.2024.1482738] [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: 08/18/2024] [Accepted: 09/25/2024] [Indexed: 10/26/2024] Open
Abstract
Despite the transformative impact of anti-PD-1/PD-L1 therapies, challenges such as low response rates persist. The stimulator of interferon genes (STING) pathway, a crucial element of innate immunity, emerges as a strategic target to overcome these limitations. Understanding its multifaceted functions in cancer, including antigen presentation and response to DNA damage, provides valuable insights. STING agonists, categorized into cyclic dinucleotides (CDNs) and non-CDNs, exhibit promising safety and efficacy profiles. Innovative delivery systems, including antibody-drug conjugates, nanocarriers, and exosome-based therapies, address challenges associated with systemic administration and enhance targeted tumor delivery. Personalized vaccines, such as DT-Exo-STING, showcase the adaptability of STING agonists for individualized treatment. These advancements not only offer new prospects for combination therapies but also pave the way for overcoming resistance mechanisms. This review focuses on the potential of targeting STING pathway to enhance cancer immunotherapy. The integration of STING agonists into cancer immunotherapy holds promise for more effective, personalized, and successful approaches against malignancies, presenting a beacon of hope for the future of cancer treatment.
Collapse
Affiliation(s)
| | - Ying Xiong
- Department of Obstetrics and Gynecology, Haiyan People’s Hospital,
Jiaxing, China
| |
Collapse
|
18
|
Tang B, Chen Y, Jiang Y, Fang M, Gao Q, Ren X, Yao L, Huang G, Chen J, Zhang X, Li R, Zhao S, Gao M, Luo R, Qi M, Li F, Zheng F, Lee M, Tao X, Duan R, Guo J, Chi Z, Cui C. Toripalimab in combination with HBM4003, an anti-CTLA-4 heavy chain-only antibody, in advanced melanoma and other solid tumors: an open-label phase I trial. J Immunother Cancer 2024; 12:e009662. [PMID: 39366752 PMCID: PMC11459314 DOI: 10.1136/jitc-2024-009662] [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: 09/08/2024] [Indexed: 10/06/2024] Open
Abstract
BACKGROUND HBM4003 is a novel anti-CTLA-4 heavy chain-only antibody, designed to enhance Treg ablation and antibody-dependent cell-mediated cytotoxicity while ensuring a manageable safety profile. This phase I trial investigated the safety, pharmacokinetics, immunogenicity and preliminary efficacy of HBM4003 plus with anti-PD-1 antibody toripalimab in patients with advanced solid tumors, especially focusing on melanoma. METHODS The multicenter, open-label phase I trial was divided into two parts: dose-escalation phase (part 1) and dose-expansion phase (part 2). In part 1, HBM4003 was administered at doses of 0.03, 0.1, 0.3 mg/kg in combination with toripalimab with fixed dosage of 240 mg every 3 weeks. The recommended phase II dose (RP2D) was used in the expansion phase. Primary endpoints were safety and RP2D in part 1 and objective response rate (ORR) in part 2. Biomarkers based on cytokines and multiplex immunofluorescence staining were explored. RESULTS A total of 40 patients received study treatment, including 36 patients treated with RP2D of HBM4003 0.3 mg/kg plus toripalimab 240 mg every 3 week. 36 participants (90.0%) experienced at least one treatment-related adverse event (TRAE), of which 10 (25.0%) patients experienced grade ≥3 TRAEs and 5 (12.5%) experienced immune-mediated adverse events (irAEs) with maximum severity of grade 3. No grade 4 or 5 irAEs occurred. Efficacy analysis set included 32 melanoma patients treated with RP2D and with available post-baseline imaging data. The ORRs of anti-PD-1/PD-L1 treatment-naïve subgroup and anti-PD-1/PD-L1 treatment-failed subgroup were 33.3% and 5.9%, respectively. In mucosal melanoma, the ORR of the two subgroups were 40.0% and 10.0%, respectively. Baseline high Treg/CD4+ratio in the tumor serves as an independent predictive factor for the efficacy of immunotherapy. CONCLUSIONS HBM4003 0.3 mg/kg plus toripalimab 240 mg every 3 week demonstrated manageable safety in solid tumors and no new safety signal. Limited data demonstrated promising antitumor activity, especially in PD-1 treatment-naïve mucosal melanoma. TRIAL REGISTRATION NUMBER NCT04727164.
Collapse
Affiliation(s)
- Bixia Tang
- Peking University Cancer Hospital & Institute, Beijing, China
| | - Yu Chen
- Department of Medical Oncology, Fujian Medical University Cancer Hospital, Fuzhou, Fujian, China
| | - Yu Jiang
- Department of Head and Neck Oncology, West China Hospital of Sichuan University, Chengdu, Sichuan Province, China
| | - Meiyu Fang
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Quanli Gao
- Immunotherapy Department, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Xiubao Ren
- Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Li Yao
- The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Gang Huang
- Central South University (Hunan Cancer Hospital), Changsha, Hunan, China
| | - Jing Chen
- Union Hospital Tongji Medical College Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaoshi Zhang
- Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Rongqing Li
- Tumor Radiotherapy Department, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | | | | | | | - Meng Qi
- Harbour BioMed, Shanghai, China
| | - Feng Li
- Harbour BioMed, Shanghai, China
| | | | | | | | - Rong Duan
- Peking University Cancer Hospital & Institute, Beijing, China
| | - Jun Guo
- Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhihong Chi
- Peking University Cancer Hospital & Institute, Beijing, China
| | - Chuanliang Cui
- Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
19
|
Verkhovskaia S, Falcone R, Di Pietro FR, Carbone ML, Samela T, Perez M, Poti G, Morelli MF, Zappalà AR, Di Rocco ZC, Morese R, Piesco G, Chesi P, Marchetti P, Abeni D, Failla CM, De Galitiis F. Survival of Patients with Metastatic Melanoma Treated with Ipilimumab after PD-1 Inhibitors: A Single-Center Real-World Study. Cancers (Basel) 2024; 16:3397. [PMID: 39410017 PMCID: PMC11475497 DOI: 10.3390/cancers16193397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/27/2024] [Accepted: 10/02/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND When monotherapy with PD-1 inhibitors in metastatic melanoma fails, there are currently no standard second-line choices. In case of the unavailability of clinical trials, ipilimumab represents a possible alternative treatment. METHODS We collected data of 44 patients who received ipilimumab after the failure of PD-1 inhibitors from July 2017 to May 2023 at our Institute. Overall survival (OS), progression-free survival (PFS), and post-progression survival (PPS) based on BRAF or NRAS mutation status, sex, and the presence of brain metastases were estimated using the Kaplan-Meier method. Cox regression was used to evaluate independence in multivariate analysis. The objective response rate (ORR) was estimated based on RECIST 1.1. RESULTS Among the 44 patients enrolled in this study, 28 BRAF-wildtype, 9 BRAF-mutated, and 7 NRAS-mutated patients were identified. OS analysis showed a significant difference between wildtype and BRAF- or NRAS-mutated patients: 23.2 months vs 5.3 and 4.59, respectively, p = 0.017. The presence of brain metastases and BRAF or NRAS mutation were independent factors for mortality in multivariate analysis. CONCLUSIONS In case of failure to enroll patients in innovative clinical trials, second-line ipilimumab still represents an effective therapy in patients with metastatic wildtype melanoma and in the absence of brain metastases.
Collapse
Affiliation(s)
- Sofia Verkhovskaia
- Department of Oncology, Istituto Dermopatico dell’Immacolata IDI-IRCCS, 00167 Rome, Italy; (S.V.); (R.F.); (F.R.D.P.); (G.P.); (M.F.M.); (A.R.Z.); (Z.C.D.R.); (R.M.); (G.P.); (P.C.); (P.M.); (F.D.G.)
| | - Rosa Falcone
- Department of Oncology, Istituto Dermopatico dell’Immacolata IDI-IRCCS, 00167 Rome, Italy; (S.V.); (R.F.); (F.R.D.P.); (G.P.); (M.F.M.); (A.R.Z.); (Z.C.D.R.); (R.M.); (G.P.); (P.C.); (P.M.); (F.D.G.)
| | - Francesca Romana Di Pietro
- Department of Oncology, Istituto Dermopatico dell’Immacolata IDI-IRCCS, 00167 Rome, Italy; (S.V.); (R.F.); (F.R.D.P.); (G.P.); (M.F.M.); (A.R.Z.); (Z.C.D.R.); (R.M.); (G.P.); (P.C.); (P.M.); (F.D.G.)
| | - Maria Luigia Carbone
- Clinical Trial Center, Istituto Dermopatico dell’Immacolata IDI-IRCCS, 00167 Rome, Italy
| | - Tonia Samela
- Epidemiology Units, Istituto Dermopatico dell’Immacolata IDI-IRCCS, 00167 Rome, Italy; (T.S.); (D.A.)
| | - Marie Perez
- Department of Histopathology, Istituto Dermopatico dell’Immacolata IDI-IRCCS, 00167 Rome, Italy;
| | - Giulia Poti
- Department of Oncology, Istituto Dermopatico dell’Immacolata IDI-IRCCS, 00167 Rome, Italy; (S.V.); (R.F.); (F.R.D.P.); (G.P.); (M.F.M.); (A.R.Z.); (Z.C.D.R.); (R.M.); (G.P.); (P.C.); (P.M.); (F.D.G.)
| | - Maria Francesca Morelli
- Department of Oncology, Istituto Dermopatico dell’Immacolata IDI-IRCCS, 00167 Rome, Italy; (S.V.); (R.F.); (F.R.D.P.); (G.P.); (M.F.M.); (A.R.Z.); (Z.C.D.R.); (R.M.); (G.P.); (P.C.); (P.M.); (F.D.G.)
| | - Albina Rita Zappalà
- Department of Oncology, Istituto Dermopatico dell’Immacolata IDI-IRCCS, 00167 Rome, Italy; (S.V.); (R.F.); (F.R.D.P.); (G.P.); (M.F.M.); (A.R.Z.); (Z.C.D.R.); (R.M.); (G.P.); (P.C.); (P.M.); (F.D.G.)
| | - Zorika Christiana Di Rocco
- Department of Oncology, Istituto Dermopatico dell’Immacolata IDI-IRCCS, 00167 Rome, Italy; (S.V.); (R.F.); (F.R.D.P.); (G.P.); (M.F.M.); (A.R.Z.); (Z.C.D.R.); (R.M.); (G.P.); (P.C.); (P.M.); (F.D.G.)
| | - Roberto Morese
- Department of Oncology, Istituto Dermopatico dell’Immacolata IDI-IRCCS, 00167 Rome, Italy; (S.V.); (R.F.); (F.R.D.P.); (G.P.); (M.F.M.); (A.R.Z.); (Z.C.D.R.); (R.M.); (G.P.); (P.C.); (P.M.); (F.D.G.)
| | - Gabriele Piesco
- Department of Oncology, Istituto Dermopatico dell’Immacolata IDI-IRCCS, 00167 Rome, Italy; (S.V.); (R.F.); (F.R.D.P.); (G.P.); (M.F.M.); (A.R.Z.); (Z.C.D.R.); (R.M.); (G.P.); (P.C.); (P.M.); (F.D.G.)
| | - Paolo Chesi
- Department of Oncology, Istituto Dermopatico dell’Immacolata IDI-IRCCS, 00167 Rome, Italy; (S.V.); (R.F.); (F.R.D.P.); (G.P.); (M.F.M.); (A.R.Z.); (Z.C.D.R.); (R.M.); (G.P.); (P.C.); (P.M.); (F.D.G.)
| | - Paolo Marchetti
- Department of Oncology, Istituto Dermopatico dell’Immacolata IDI-IRCCS, 00167 Rome, Italy; (S.V.); (R.F.); (F.R.D.P.); (G.P.); (M.F.M.); (A.R.Z.); (Z.C.D.R.); (R.M.); (G.P.); (P.C.); (P.M.); (F.D.G.)
| | - Damiano Abeni
- Epidemiology Units, Istituto Dermopatico dell’Immacolata IDI-IRCCS, 00167 Rome, Italy; (T.S.); (D.A.)
| | - Cristina Maria Failla
- Experimental Immunology Laboratory, Istituto Dermopatico dell’Immacolata IDI-IRCCS, 00167 Rome, Italy;
| | - Federica De Galitiis
- Department of Oncology, Istituto Dermopatico dell’Immacolata IDI-IRCCS, 00167 Rome, Italy; (S.V.); (R.F.); (F.R.D.P.); (G.P.); (M.F.M.); (A.R.Z.); (Z.C.D.R.); (R.M.); (G.P.); (P.C.); (P.M.); (F.D.G.)
| |
Collapse
|
20
|
Sari G, Dhatchinamoorthy K, Orellano-Ariza L, Ferreira LM, Brehm MA, Rock K. IRF2 loss is associated with reduced MHC I pathway transcripts in subsets of most human cancers and causes resistance to checkpoint immunotherapy in human and mouse melanomas. J Exp Clin Cancer Res 2024; 43:276. [PMID: 39354629 PMCID: PMC11446056 DOI: 10.1186/s13046-024-03187-5] [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: 06/25/2024] [Accepted: 09/07/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND In order for cancers to progress, they must evade elimination by CD8 T cells or other immune mechanisms. CD8 T cells recognize and kill tumor cells that display immunogenic tumor peptides bound to MHC I molecules. One of the ways that cancers can escape such killing is by reducing expression of MHC I molecules, and loss of MHC I is frequently observed in tumors. There are multiple different mechanisms that can underly the loss of MHC I complexes on tumor and it is currently unclear whether there are particular mechanisms that occur frequently and, if so, in what types of cancers. Also of importance to know is whether the loss of MHC I is reversible and how such loss and/or its restoration would impact responses to immunotherapy. Here, we investigate these issues for loss of IRF1 and IRF2, which are transcription factors that drive expression of MHC I pathway genes and some killing mechanisms. METHODS Bioinformatics analyses of IRF2 and IRF2-dependent gene transcripts were performed for all human cancers in the TCGA RNAseq database. IRF2 protein-DNA-binding was analyzed in ChIPseq databases. CRISRPcas9 was used to knock out IRF1 and IRF2 genes in human and mouse melanoma cells and the resulting phenotypes were analyzed in vitro and in vivo. RESULTS Transcriptomic analysis revealed that IRF2 expression was reduced in a substantial subset of cases in almost all types of human cancers. When this occurred there was a corresponding reduction in the expression of IRF2-regulated genes that were needed for CD8 T cell recognition. To test cause and effect for these IRF2 correlations and the consequences of IRF2 loss, we gene-edited IRF2 in a patient-derived melanoma and a mouse melanoma. The IRF2 gene-edited melanomas had reduced expression of transcripts for genes in the MHC I pathway and decreased levels of MHC I complexes on the cell surface. Levels of Caspase 7, an IRF2 target gene involved in CD8 T cell killing of tumors, were also reduced. This loss of IRF2 caused both human and mouse melanomas to become resistant to immunotherapy with a checkpoint inhibitor. Importantly, these effects were reversible. Stimulation of the IRF2-deficient melanomas with interferon induced the expression of a functionally homologous transcription factor, IRF1, which then restored the MHC I pathway and responsiveness to CPI. CONCLUSIONS Our study shows that a subset of cases within most types of cancers downregulates IRF2 and that this can allow cancers to escape immune control. This can cause resistance to checkpoint blockade immunotherapy and is reversible with currently available biologics.
Collapse
Affiliation(s)
- G Sari
- Department of Pathology, UMass Chan Medical School, 55 Lake Avenue North, Worcester, MA, 01655, USA
| | - K Dhatchinamoorthy
- Department of Pathology, UMass Chan Medical School, 55 Lake Avenue North, Worcester, MA, 01655, USA
| | - L Orellano-Ariza
- Department of Pathology, UMass Chan Medical School, 55 Lake Avenue North, Worcester, MA, 01655, USA
| | - L M Ferreira
- Program in Molecular Medicine, Diabetes Center of Excellence, UMass Chan Medical School, Worcester, MA, USA
| | - M A Brehm
- Program in Molecular Medicine, Diabetes Center of Excellence, UMass Chan Medical School, Worcester, MA, USA
| | - K Rock
- Department of Pathology, UMass Chan Medical School, 55 Lake Avenue North, Worcester, MA, 01655, USA.
| |
Collapse
|
21
|
Martín-Lluesma S, Svane IM, Dafni U, Vervita K, Karlis D, Dimopoulou G, Tsourti Z, Rohaan MW, Haanen JBAG, Coukos G. Efficacy of TIL therapy in advanced cutaneous melanoma in the current immuno-oncology era: updated systematic review and meta-analysis. Ann Oncol 2024; 35:860-872. [PMID: 39053767 DOI: 10.1016/j.annonc.2024.07.723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 06/06/2024] [Accepted: 07/08/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND Adoptive cell therapy with tumor-infiltrating lymphocytes (TIL-ACT) has consistently shown efficacy in advanced melanoma. New results in the field provide now the opportunity to assess overall survival (OS) after TIL-ACT and to examine the effect of prior anti-programmed cell death protein 1/programmed death-ligand 1 [anti-PD-(L)1] therapy on its efficacy. METHODS A comprehensive search was conducted in PubMed up to 29 February 2024. Ιn this meta-analysis we focused on studies including high-dose interleukin 2, doubling the patient numbers from our previous meta-analysis conducted up to December 2018 and using OS as the primary endpoint. Objective response rate (ORR), complete response rate (CRR), and duration of response were secondary endpoints. Findings are synthesized using tables, Kaplan-Meier plots, and forest plots. Pooled estimates for ORR and CRR were derived from fixed or random effects models. RESULTS A total of 13 high-dose interleukin 2 studies were included in this updated meta-analysis, with OS information available for 617 patients. No difference was found in median OS between studies with prior anti-PD-(L)1 treatment {n = 238; 17.5 months [95% confidence interval (CI) 13.8-20.5 months]} and without [n = 379; 16.3 months (95% CI 14.2-20.6 months)] (log-rank P = 0.53). ORR was estimated to be 34% (95% CI 16%-52%) and 44% (95% CI 37%-51%), for the studies with and without prior anti-PD-(L)1, respectively. The pooled estimate for CRR was 10% for both groups. No statistically significant difference was observed between the two groups, either for ORR (P = 0.15) or CRR (P = 0.45). CONCLUSIONS Prior anti-PD-(L)1 treatment has no effect on the clinical response or survival benefit from TIL-ACT in advanced cutaneous melanoma. The benefit of TIL therapy in the second-line setting is also present after anti-PD-(L)1 treatment. Our data reinforce the evidence that TIL-ACT should be considered as a treatment of choice in second line for metastatic melanoma patients failing anti-PD-(L)1 therapy.
Collapse
Affiliation(s)
- S Martín-Lluesma
- Department of Medical Oncology, Vall d'Hebron University Hospital, Barcelona; Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - I M Svane
- Department of Oncology, National Center for Cancer Immune Therapy (CCIT-DK), Copenhagen University Hospital, Herlev, Denmark
| | - U Dafni
- Faculty of Nursing, National and Kapodistrian University of Athens, Athens, Greece; Department of Oncology, CHUV, University of Lausanne, Lausanne, Switzerland.
| | - K Vervita
- Scientific Research Consulting Hellas, Statistics Center, Athens
| | - D Karlis
- Department of Statistics, Athens University of Economics and Business, Athens, Greece
| | - G Dimopoulou
- Scientific Research Consulting Hellas, Statistics Center, Athens
| | - Z Tsourti
- Scientific Research Consulting Hellas, Statistics Center, Athens
| | - M W Rohaan
- Division of Medical Oncology, Netherlands Cancer Institute (NKI), Amsterdam
| | - J B A G Haanen
- Division of Medical Oncology, Netherlands Cancer Institute (NKI), Amsterdam; Department of Medical Oncology, Leiden University Medical Oncology, Leiden, Netherlands; Melanoma Clinic, CHUV, Lausanne
| | - G Coukos
- Department of Oncology, Lausanne University Hospital and University of Lausanne Ludwig Institute for Cancer Research Lausanne Branch, Switzerland
| |
Collapse
|
22
|
Gonzalez-Cao M, Puertolas T, Manzano JL, Maldonado C, Yelamos O, Berciano-Guerrero MÁ, Cerezuela P, Martin-Liberal J, Muñoz-Couselo E, Espinosa E, Drozdowskyj A, Berrocal A, Soria A, Marquez-Rodas I, Martin-Algarra S, Quindos M, Puig S. Access to melanoma drugs in Spain: a cross-sectional survey. Clin Transl Oncol 2024; 26:2572-2583. [PMID: 38750345 DOI: 10.1007/s12094-024-03501-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 04/17/2024] [Indexed: 09/20/2024]
Abstract
BACKGROUND The development of highly active drugs has improved the survival of melanoma patients, but elevated drug prices place a significant burden on health care systems. In Spain, the public health care system is transferred to the 17 autonomous communities (AACC). The objective of this study is to describe the situation of drug access for melanoma patients in Spain and how this decentralized system is affecting equity. METHODS From July to September 2023, a cross-sectional survey was sent to members of the Spanish Multidisciplinary Melanoma Group (GEM Group). The questionnaire consulted about the real access to new drugs in each hospital. The responses were collected anonymously and analyzed according to several variables, including the AACC. RESULTS The survey was answered by 50 physicians in 15 AACC. No major differences on access between AACC were observed for indications that are reimbursed by the Spanish Health Care System (adjuvant immunotherapy for stage IIIC-IIID and resected stage IV melanoma). Important differences in drug access were observed among AACC and among centers within the same AACC, for most of the EMA indications that are not reimbursed (adjuvant immunotherapy for stages IIB-IIC-IIIA-IIIB) or that are not fully reimbursed (ipilimumab plus nivolumab in advanced stage). Homogeneously, access to adjuvant targeted drugs, TIL therapy and T-VEC, is extremely low or non-existing in all AACC. CONCLUSIONS For most indications that reimbursement is restricted out of the EMA indication, a great diversity on access was found throughout the different hospitals in Spain, including heterogeneity intra-AACC.
Collapse
Affiliation(s)
- Maria Gonzalez-Cao
- Translational Cancer Research Unit, Instituto Oncologico Dr Rosell, Dexeus University Hospital, C/Sabino Arana, 5, 080028, Barcelona, Spain.
| | | | - Jose Luis Manzano
- Oncology Department, Hospital Germans Trias I Pujol, Badalona, Spain
| | - Cayetana Maldonado
- Dermatology Department, Hospital Universitario de Asturias, Oviedo, Spain
| | - Oriol Yelamos
- Dermatology Department, Hospital de La Santa Creu I Sant Pau, Barcelona, Spain
| | - Miguel Ángel Berciano-Guerrero
- Oncology Department Hospitales, Universitarios Regional y Virgen de La Victoria (HURyVV), Instituto de Investigaciones Biomédicas de Málaga (IBIMA), Málaga, Spain
| | - Pablo Cerezuela
- Oncology Department, Hospital Clínico Universitario Virgen de La Arrixaca, Instituto Murciano de Investigación Biosanitaria (IMIB), Ciudad de Murcia, Murcia, Spain
| | | | | | - Enrique Espinosa
- Oncology Department, Hospital Universitario La Paz, Madrid, Spain
- CIBERER, Barcelona, Spain
| | - Ana Drozdowskyj
- Translational Cancer Research Unit, Instituto Oncologico Dr Rosell, Dexeus University Hospital, C/Sabino Arana, 5, 080028, Barcelona, Spain
| | - Alfonso Berrocal
- Oncology Department, Hospital General de Valencia, Valencia, Spain
| | - Ainara Soria
- Oncology Department, Hospital Ramon y Cajal, Madrid, Spain
| | - Ivan Marquez-Rodas
- Oncology Department, Hospital General Universitario Gregorio Marañon, Madrid, Spain
| | | | - Maria Quindos
- Medical Oncology Department, Complexo Hospitalario Universitario de A Coruña. Biomedical Research Institute (INIBIC), A Coruña, Spain
| | - Susana Puig
- CIBERER, Barcelona, Spain.
- Dermatology Department, Hospital Clinic Barcelona, University of Barcelona, IDIBAPS, C Villrroel, 08023, Barcelona, Spain.
| |
Collapse
|
23
|
Mao Z, Hu Y, Zhao Y, Zhang X, Guo L, Wang X, Zhang J, Miao M. The Mutual Regulatory Role of Ferroptosis and Immunotherapy in Anti-tumor Therapy. Apoptosis 2024; 29:1291-1308. [PMID: 38853203 PMCID: PMC11416416 DOI: 10.1007/s10495-024-01988-9] [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: 05/25/2024] [Indexed: 06/11/2024]
Abstract
Ferroptosis is a form of cell death that is triggered by the presence of ferrous ions and is characterized by lipid peroxidation induced by these ions. The mechanism exhibits distinct morphological characteristics compared to apoptosis, autophagy, and necrosis. A notable aspect of ferroptosis is its ability to inhibit uncontrolled tumor replication and immortalization, especially in malignant, drug-resistant, and metastatic tumors. Additionally, immunotherapy, a novel therapeutic approach for tumors, has been found to have a reciprocal regulatory relationship with ferroptosis in the context of anti-tumor therapy. A comprehensive analysis of ferroptosis and immunotherapy in tumor therapy is presented in this paper, highlighting the potential for mutual adjuvant effects. Specifically, we discuss the mechanisms underlying ferroptosis and immunotherapy, emphasizing their ability to improve the tumor immune microenvironment and enhance immunotherapeutic effects. Furthermore, we investigate how immunotherapeutic factors may increase the sensitivity of tumor cells to ferroptosis. We aim to provide a prospective view of the promising value of combined ferroptosis and immunotherapy in anticancer therapy by elucidating the mutual regulatory network between each.
Collapse
Affiliation(s)
- Zhiguo Mao
- Department of Pharmacology, Zhengdong New District, Henan University of Chinese Medicine, No. 156 Jinshui East Road, Zhengzhou, 450046, Henan, China
- Collaborative Innovation Center of Research and Development, Whole Industry Chain of Yu-Yao in Henan Province, Henan, China
| | - Yilong Hu
- Department of Pharmacology, Zhengdong New District, Henan University of Chinese Medicine, No. 156 Jinshui East Road, Zhengzhou, 450046, Henan, China
- Collaborative Innovation Center of Research and Development, Whole Industry Chain of Yu-Yao in Henan Province, Henan, China
| | - Yinan Zhao
- Department of Pharmacology, Zhengdong New District, Henan University of Chinese Medicine, No. 156 Jinshui East Road, Zhengzhou, 450046, Henan, China
- Collaborative Innovation Center of Research and Development, Whole Industry Chain of Yu-Yao in Henan Province, Henan, China
| | - Xiaolei Zhang
- Department of Pharmacology, Zhengdong New District, Henan University of Chinese Medicine, No. 156 Jinshui East Road, Zhengzhou, 450046, Henan, China
- Collaborative Innovation Center of Research and Development, Whole Industry Chain of Yu-Yao in Henan Province, Henan, China
| | - Lin Guo
- Department of Pharmacology, Zhengdong New District, Henan University of Chinese Medicine, No. 156 Jinshui East Road, Zhengzhou, 450046, Henan, China
- Collaborative Innovation Center of Research and Development, Whole Industry Chain of Yu-Yao in Henan Province, Henan, China
| | - Xiaoran Wang
- Department of Pharmacology, Zhengdong New District, Henan University of Chinese Medicine, No. 156 Jinshui East Road, Zhengzhou, 450046, Henan, China
- Collaborative Innovation Center of Research and Development, Whole Industry Chain of Yu-Yao in Henan Province, Henan, China
| | - Jinying Zhang
- Department of Pharmacology, Zhengdong New District, Henan University of Chinese Medicine, No. 156 Jinshui East Road, Zhengzhou, 450046, Henan, China
- Collaborative Innovation Center of Research and Development, Whole Industry Chain of Yu-Yao in Henan Province, Henan, China
| | - Mingsan Miao
- Department of Pharmacology, Zhengdong New District, Henan University of Chinese Medicine, No. 156 Jinshui East Road, Zhengzhou, 450046, Henan, China.
- Collaborative Innovation Center of Research and Development, Whole Industry Chain of Yu-Yao in Henan Province, Henan, China.
| |
Collapse
|
24
|
Xu F, Ni Q, Gong N, Xia B, Zhang J, Guo W, Hu Z, Li J, Liang XJ. Delivery Systems Developed for Treatment Combinations to Improve Adoptive Cell Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2407525. [PMID: 39165065 DOI: 10.1002/adma.202407525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/26/2024] [Indexed: 08/22/2024]
Abstract
Adoptive cell therapy (ACT) has shown great success in the clinic for treating hematologic malignancies. However, solid tumor treatment with ACT monotherapy is still challenging, owing to insufficient expansion and rapid exhaustion of adoptive cells, tumor antigen downregulation/loss, and dense tumor extracellular matrix. Delivery strategies for combination cell therapy have great potential to overcome these hurdles. The delivery of vaccines, immune checkpoint inhibitors, cytokines, chemotherapeutics, and photothermal reagents in combination with adoptive cells, have been shown to improve the expansion/activation, decrease exhaustion, and promote the penetration of adoptive cells in solid tumors. Moreover, the delivery of nucleic acids to engineer immune cells directly in vivo holds promise to overcome many of the hurdles associated with the complex ex vivo cell engineering strategies. Here, these research advance, as well as the opportunities and challenges for integrating delivery technologies into cell therapy s are discussed, and the outlook for these emerging areas are criticlly analyzed.
Collapse
Affiliation(s)
- Fengfei Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Qiankun Ni
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
- Department of Chemistry, Center for BioAnalytical Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, New Cornerstone Science Institute, Tsinghua University, Beijing, China
| | - Ningqiang Gong
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Bozhang Xia
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jinchao Zhang
- College of Chemistry & Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, State Key Laboratory of New Pharmaceutical Preparations and Excipients, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, China
| | - Weisheng Guo
- College of Biomedical Engineering, Guangzhou Medical University, Guangzhou, 510260, China
| | - Zhongbo Hu
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jinghong Li
- Department of Chemistry, Center for BioAnalytical Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, New Cornerstone Science Institute, Tsinghua University, Beijing, China
| | - Xing-Jie Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
25
|
Robinson MA, Kennedy A, Orozco CT, Chen HC, Waters E, Giovacchini D, Yeung K, Filer L, Hinze C, Lloyd C, Dovedi SJ, Sansom DM. Rigid, bivalent CTLA-4 binding to CD80 is required to disrupt the cis CD80/PD-L1 interaction. Cell Rep 2024; 43:114768. [PMID: 39277860 DOI: 10.1016/j.celrep.2024.114768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 08/20/2024] [Accepted: 08/30/2024] [Indexed: 09/17/2024] Open
Abstract
The CTLA-4 and PD-1 checkpoints control immune responses and are key targets in immunotherapy. Both pathways are connected via a cis interaction between CD80 and PD-L1, the ligands for CTLA-4 and PD-1, respectively. This cis interaction prevents PD-1-PD-L1 binding but is reversed by CTLA-4 trans-endocytosis of CD80. However, how CTLA-4 selectively removes CD80, but not PD-L1, is unclear. Here, we show CTLA-4-CD80 interactions are unimpeded by PD-L1 and that CTLA-4 binding with CD80 does not displace PD-L1 per se. Rather, both rigidity and bivalency of CTLA-4 molecules are required to orientate CD80 such that PD-L1 interactions are no longer permissible. Moreover, soluble CTLA-4 released PD-L1 only at specific expression levels of CD80 and PD-L1, whereas CTLA-4 trans-endocytosis released PD-L1 in all conditions. These data show that PD-L1 release from CD80 is driven by orientation and bivalent cross-linking of membrane proteins and that trans-endocytosis of CD80 efficiently promotes PD-L1 availability.
Collapse
Affiliation(s)
- Maximillian A Robinson
- Institute of Immunity and Transplantation, Pears Building, Rowland Hill St, London NW3 2PP, UK
| | - Alan Kennedy
- Institute of Immunity and Transplantation, Pears Building, Rowland Hill St, London NW3 2PP, UK
| | - Carolina T Orozco
- Biologics Engineering, R&D, AstraZeneca, 1, Francis Crick Avenue Cambridge CB2 0AA, UK
| | - Hung-Chang Chen
- Early Oncology ICC, R&D, AstraZeneca, 1, Francis Crick Avenue, Cambridge CB2 0AA, UK
| | - Erin Waters
- Institute of Immunity and Transplantation, Pears Building, Rowland Hill St, London NW3 2PP, UK
| | - Dalisay Giovacchini
- Institute of Immunity and Transplantation, Pears Building, Rowland Hill St, London NW3 2PP, UK
| | - Kay Yeung
- Institute of Immunity and Transplantation, Pears Building, Rowland Hill St, London NW3 2PP, UK
| | - Lily Filer
- Institute of Immunity and Transplantation, Pears Building, Rowland Hill St, London NW3 2PP, UK
| | - Claudia Hinze
- Institute of Immunity and Transplantation, Pears Building, Rowland Hill St, London NW3 2PP, UK
| | - Christopher Lloyd
- Biologics Engineering, R&D, AstraZeneca, 1, Francis Crick Avenue Cambridge CB2 0AA, UK
| | - Simon J Dovedi
- Early Oncology ICC, R&D, AstraZeneca, 1, Francis Crick Avenue, Cambridge CB2 0AA, UK
| | - David M Sansom
- Institute of Immunity and Transplantation, Pears Building, Rowland Hill St, London NW3 2PP, UK.
| |
Collapse
|
26
|
Okuda-Hiwatashi S, Amagai R, Fujimura T, Kambayashi Y, Watanabe-Takahashi M, Yamazaki E, Tamabuchi E, Itabashi C, Hashimoto A, Asano Y. The Evaluation of Immune Checkpoint Inhibitors and BRAF/MEK Inhibitors in Different Therapy Lines for Metastatic Melanoma: A Retrospective Study. J Clin Med 2024; 13:5560. [PMID: 39337055 PMCID: PMC11432506 DOI: 10.3390/jcm13185560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/18/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Background: Nivolumab plus ipilimumab (nivo/ipi) combination therapy is highly effective in treating advanced melanoma, but serious immune-related adverse events (irAEs) are prevalent. The overall response rate (ORR) of the BRAF inhibitor plus MEK inhibitor (BRAFi/MEKi) combination therapy for BRAFV600-mutant advanced melanoma surpasses that of immune checkpoint inhibitors (ICIs). However, the OS and PFS of BRAFi/MEKi combination therapy are inferior to those of ICIs. Methods: We retrospectively evaluated 22 melanoma patients treated with nivo/ipi therapy and 13 patients treated with encorafenib plus binimetinib (enco/bini) between November 2018 and July 2023. Results: The ORR of nivo/ipi for metastatic melanoma patients was significantly higher in the first-line cohort [60.0% (95% CI: 31.2-83.3%)] than in the second-line or beyond cohort [8.3% (95% CI: 0-37.5%)], whereas the ORR of enco/bini was comparable between the first-line cohort [75.0% (95% CI: 28.9-96.6%)] and the second-line or beyond cohort [77.8% (95% CI: 44.3-94.7%)]. The median PFS of nivo/ipi significantly improved in the first-line cohort [7.7 months (95% CI: 2.0-11.9)] compared to the second-line or beyond cohort [2.3 months (95% CI: 0.5-6.0)] (p = 0.0109). In addition to efficacy, the incidence of grade 3 or greater AEs was comparable in the first-line and second-line or beyond cohorts. Conclusions: Although our present data are based on a small number of cases, they suggest that nivo/ipi should be administered as the first-line therapy for the treatment of BRAFV600-mutant metastatic melanoma, rather than enco/bini, aligning with findings from previous clinical trials.
Collapse
Affiliation(s)
| | | | - Taku Fujimura
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan; (S.O.-H.); (R.A.); (Y.K.); (M.W.-T.); (E.Y.); (E.T.); (C.I.); (A.H.); (Y.A.)
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Sari G, Dhatchinamoorthy K, Orellano-Ariza L, Ferreira LM, Brehm MA, Rock K. IRF2 loss is associated with reduced MHC I pathway transcripts in subsets of most human cancers and causes resistance to checkpoint immunotherapy in human and mouse melanomas. RESEARCH SQUARE 2024:rs.3.rs-4997954. [PMID: 39281881 PMCID: PMC11398557 DOI: 10.21203/rs.3.rs-4997954/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
Background In order for cancers to progress, they must evade elimination by CD8 T cells or other immune mechanisms. CD8 T cells recognize and kill tumor cells that display immunogenic tumor peptides bound to MHC I molecules. One of the ways that cancers can escape such killing is by reducing expression of MHC I molecules, and loss of MHC I is frequently observed in tumors. There are multiple different mechanisms that can underly the loss of MHC I complexes on tumor and it is currently unclear whether there are particular mechanisms that occur frequently and, if so, in what types of cancers. Also of importance to know is whether the loss of MHC I is reversible and how such loss and/or its restoration would impact responses to immunotherapy. Here, we investigate these issues for loss of IRF1 and IRF2, which are transcription factors that drive expression of MHC I pathway genes and some killing mechanisms. Methods Bioinformatics analyses of IRF2 and IRF2-dependent gene transcripts were performed for all human cancers in the TCGA RNAseq database. IRF2 protein-DNA-binding was analyzed in ChIPseq databases. CRISRPcas9 was used to knock out IRF1 and IRF2 genes in human and mouse melanoma cells and the resulting phenotypes were analyzed in vitro and in vivo. Results Transcriptomic analysis revealed that IRF2 expression was reduced in a substantial subset of cases in almost all types of human cancers. When this occurred there was a corresponding reduction in the expression of IRF2-regulated genes that were needed for CD8 T cell recognition. To test cause and effect for these IRF2 correlations and the consequences of IRF2 loss, we gene-edited IRF2 in a patient-derived melanoma and a mouse melanoma. The IRF2 gene-edited melanomas had reduced expression of transcripts for genes in the MHC I pathway and decreased levels of MHC I complexes on the cell surface. Levels of Caspase 7, an IRF2 target gene involved in CD8 T cell killing of tumors, were also reduced. This loss of IRF2 caused both human and mouse melanomas to become resistant to immunotherapy with a checkpoint inhibitor. Importantly, these effects were reversible. Stimulation of the IRF2-deficient melanomas with interferon induced the expression of a functionally homologous transcription factor, IRF1, which then restored the MHC I pathway and responsiveness to CPI. Conclusions Our study shows that a subset of cases within most types of cancers downregulates IRF2 and that this can allow cancers to escape immune control. This can cause resistance to checkpoint blockade immunotherapy and is reversible with currently available biologics.
Collapse
Affiliation(s)
- Gulce Sari
- Department of Pathology, UMass Chan Medical School, Worcester, MA, USA
| | | | | | - Lindsay M Ferreira
- Program in Molecular Medicine, Diabetes Center of Excellence, UMass Chan Medical School, Worcester, MA, USA
| | - Michael A Brehm
- Program in Molecular Medicine, Diabetes Center of Excellence, UMass Chan Medical School, Worcester, MA, USA Kenneth Rock
| | - Kenneth Rock
- Department of Pathology, UMass Chan Medical School, Worcester, MA, USA
| |
Collapse
|
28
|
Markussen A, Johansen JS, Larsen FO, Theile S, Hasselby JP, Willemoe GL, Lorentzen T, Madsen K, Høgdall E, Poulsen TS, Wilken EE, Geertsen P, Behrens CP, Svane IM, Nielsen D, Chen IM. Nivolumab with or without Ipilimumab Combined with Stereotactic Body Radiotherapy in Patients with Metastatic Biliary Tract Cancer: A Randomized Phase 2 Study. Clin Cancer Res 2024; 30:3428-3437. [PMID: 38874506 DOI: 10.1158/1078-0432.ccr-24-0286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 04/02/2024] [Accepted: 06/11/2024] [Indexed: 06/15/2024]
Abstract
PURPOSE The purpose of this study was to evaluate the clinical benefits of nivolumab with/without ipilimumab combined with stereotactic body radiotherapy (SBRT) in patients with pretreated metastatic biliary tract cancer (mBTC). PATIENTS AND METHODS The study was a phase 2 randomized trial with Simon's optimal two-stage design requiring 36 evaluable patients per group after second stage. Sixty-one patients were included from September 2018 to January 2022 and randomized (1:1) to receive SBRT (15 Gy × 1 on day 1 to a primary or metastatic lesion) and nivolumab (3 mg/kg intravenously on day 1 and every 2 weeks) with/without ipilimumab (1 mg/kg intravenously on day 1 and every 6 weeks). Primary endpoint was clinical benefit rate (CBR), defined as the percentage of patients with complete response, partial response, or stable disease. Decision to continue accrual into the second stage depended on the CBR from the first stage. RESULTS Forty-two patients received SBRT/nivolumab/ipilimumab with a CBR of 31.0% [95% confidence interval (CI), 17.6-47.1]. Five patients (11.9%) achieved partial response with median duration of 4.4 months (range, 1.1-21.5). Nineteen patients received SBRT/nivolumab. This group was closed after the initial stage based on a CBR of 10.5% (95% CI, 1.3-33.1). Adverse events were graded with National Cancer Institute Common Terminology Criteria for Adverse Events version 4.0. Grade ≥3 treatment-related adverse events occurred in 13 (31%) and 3 (16%) patients in the SBRT/nivolumab/ipilimumab and SBRT/nivolumab groups, respectively. One patient died from immune-related hepatitis in the SBRT/nivolumab/ipilimumab group. CONCLUSIONS Combining SBRT, nivolumab, and ipilimumab is well tolerated, feasible, and shows response in a subgroup of patients with mBTC.
Collapse
Affiliation(s)
- Alice Markussen
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
| | - Julia S Johansen
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- Department of Medicine, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, Copenhagen University, Copenhagen, Denmark
| | - Finn O Larsen
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
| | - Susann Theile
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
| | - Jane P Hasselby
- Department of Pathology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Gro L Willemoe
- Department of Pathology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Torben Lorentzen
- Department of Gastroenterology, Unit of Surgical Ultrasound, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
| | - Kasper Madsen
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
| | - Estrid Høgdall
- Department of Pathology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
| | - Tim S Poulsen
- Department of Pathology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
| | - Eva E Wilken
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
| | - Poul Geertsen
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
| | - Claus P Behrens
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- Department of Health Technology, Technical University of Denmark, Roskilde, Denmark
| | - Inge M Svane
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, Copenhagen University, Copenhagen, Denmark
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
| | - Dorte Nielsen
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, Copenhagen University, Copenhagen, Denmark
| | - Inna M Chen
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
| |
Collapse
|
29
|
Li G, Liu X, Gu C, Ma G, Li S, Ma Z, Xiong Y, Jiang Y, Huang Q, Wu J, Wu Z, Liao W, Wu Q, Shi M. Mutual exclusivity and co-occurrence patterns of immune checkpoints indicate NKG2A relates to anti-PD-1 resistance in gastric cancer. J Transl Med 2024; 22:718. [PMID: 39097734 PMCID: PMC11298088 DOI: 10.1186/s12967-024-05503-1] [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: 05/14/2024] [Accepted: 07/11/2024] [Indexed: 08/05/2024] Open
Abstract
BACKGROUND An increasing number of clinical studies have begun to explore combination strategies with immune checkpoint inhibitors, aiming to present new opportunities for overcoming anti-PD-1 treatment resistance in gastric cancer. Unfortunately, the exploration of certain immune checkpoint inhibitor combination strategies has yielded suboptimal results. Therefore, it is necessary to comprehensively analyze the expression patterns of immune checkpoints and identify optimal combination regimens of anti-PD-1 inhibitors with other immune checkpoint inhibitors. METHODS Leveraging single-cell RNA sequencing (scRNA-seq) and multivariate linear regression interaction models, we dissected the immune checkpoint expression characteristics of CD8+ T cells in gastric cancer and the immune checkpoint expression pattern (ICEP) mediating anti-PD-1 treatment resistance. Furthermore, we employed transcription factor analysis and CellOracle to explore the transcriptional regulatory mechanisms governing CD8+ T cell differentiation fates. Finally, we utilized Nichenet and spatial transcriptomic analysis to investigate the spatial expression patterns of immune checkpoints. RESULTS Interaction analysis indicated that, among the known immune checkpoints, co-expression of NKG2A and PD-1 might exert a more profound inhibitory effect on the proliferative capacity of CD8+ T cells. The co-expression analysis revealed differential co-expression pattern of PD-1 and NKG2A, defined as ICEP1 (CD8+ T cells co-expressing PD-1, CTLA-4, TIGIT, LAG-3 or CD38) and ICEP2 (CD8+ T cells solely expressing NKG2A or co-expressing with other immune checkpoints), reflecting the co-occurrence pattern of PD-1 and the mutual exclusivity of NKG2A. Further, these two ICEP CD8+ T cell subsets represented distinct CD8+ T cell differentiation fates governed by MSC and RUNX3. Notably, ICEP2 CD8+ T cells were associated with anti-PD-1 therapy resistance in gastric cancer. This phenomenon may be attributed to the recruitment of LGMN+ macrophages mediated by the CXCL16-CXCR6 signaling pathway. CONCLUSION This study unveiled two distinct ICEPs and the mutually exclusivity and co-occurrence characteristics of CD8+ T cells in gastric cancer. The ICEP2 CD8+ T cell subset, highly expressed in gastric cancer patients resistant to anti-PD-1 therapy, may be recruited by LGMN+ macrophages through CXCL16-CXCR6 axis. These findings provide evidence for NKG2A as a novel immunotherapeutic target in gastric cancer and offer new insights into combination strategies for immune checkpoint inhibitors in gastric cancer.
Collapse
Affiliation(s)
- Guanjun Li
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Xiaohan Liu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Chunhui Gu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Guichuang Ma
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Shaowei Li
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Zhenfeng Ma
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Yanqiu Xiong
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Yu Jiang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Qiong Huang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Jianhua Wu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Zhenzhen Wu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Qijing Wu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| | - Min Shi
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
30
|
Zhou Q, Jin X, Zhao Y, Wang Y, Tao M, Cao Y, Yin X. Melanoma-associated fibroblasts in tumor-promotion flammation and antitumor immunity: novel mechanisms and potential immunotherapeutic strategies. Hum Mol Genet 2024; 33:1186-1193. [PMID: 38538564 PMCID: PMC11190611 DOI: 10.1093/hmg/ddae056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 03/09/2024] [Accepted: 03/14/2024] [Indexed: 06/22/2024] Open
Abstract
Melanoma, renowned for its aggressive behavior and resistance to conventional treatments, stands as a formidable challenge in the oncology landscape. The dynamic and complex interplay between cancer cells and the tumor microenvironment has gained significant attention, revealing Melanoma-Associated Fibroblasts (MAFs) as central players in disease progression. The heterogeneity of MAFs endows them with a dual role in melanoma. This exhaustive review seeks to not only shed light on the multifaceted roles of MAFs in orchestrating tumor-promoting inflammation but also to explore their involvement in antitumor immunity. By unraveling novel mechanisms underlying MAF functions, this review aims to provide a comprehensive understanding of their impact on melanoma development. Additionally, it delves into the potential of leveraging MAFs for innovative immunotherapeutic strategies, offering new avenues for enhancing treatment outcomes in the challenging realm of melanoma therapeutics.
Collapse
Affiliation(s)
- Qiujun Zhou
- Department of Dermatology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), #54 Youdian Road, Shangcheng District, Hangzhou, Zhejiang 310000, China
- Department of First Clinical Medical College, Zhejiang Chinese Medical University, #548 Binwen Road, Binjiang District, Hangzhou, Zhejiang 310000, China
| | - Xiaoliang Jin
- Department of Dermatology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), #54 Youdian Road, Shangcheng District, Hangzhou, Zhejiang 310000, China
- Department of First Clinical Medical College, Zhejiang Chinese Medical University, #548 Binwen Road, Binjiang District, Hangzhou, Zhejiang 310000, China
| | - Ying Zhao
- Department of Dermatology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), #54 Youdian Road, Shangcheng District, Hangzhou, Zhejiang 310000, China
- Department of First Clinical Medical College, Zhejiang Chinese Medical University, #548 Binwen Road, Binjiang District, Hangzhou, Zhejiang 310000, China
| | - Yueping Wang
- Department of Dermatology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), #54 Youdian Road, Shangcheng District, Hangzhou, Zhejiang 310000, China
- Department of First Clinical Medical College, Zhejiang Chinese Medical University, #548 Binwen Road, Binjiang District, Hangzhou, Zhejiang 310000, China
| | - Maocan Tao
- Department of Dermatology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), #54 Youdian Road, Shangcheng District, Hangzhou, Zhejiang 310000, China
| | - Yi Cao
- Department of Dermatology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), #54 Youdian Road, Shangcheng District, Hangzhou, Zhejiang 310000, China
| | - Xiaohu Yin
- Department of Dermatology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), #54 Youdian Road, Shangcheng District, Hangzhou, Zhejiang 310000, China
| |
Collapse
|
31
|
Cao Y, Tang L, Fu C, Yin Y, Liu H, Feng J, Gao J, Shu W, Li Z, Zhu Y, Wang W. Black Phosphorus Quantum Dot Loaded Bioinspired Nanoplatform Synergized with aPD-L1 for Multimode Cancer Immunotherapy. NANO LETTERS 2024; 24:6767-6777. [PMID: 38771956 DOI: 10.1021/acs.nanolett.4c01511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2024]
Abstract
Efforts to prolong the blood circulation time and bypass immune clearance play vital roles in improving the therapeutic efficacy of nanoparticles (NPs). Herein, a multifunctional nanoplatform (BPP@RTL) that precisely targets tumor cells is fabricated by encapsulating ultrasmall phototherapeutic agent black phosphorus quantum dot (BPQD), chemotherapeutic drug paclitaxel (PTX), and immunomodulator PolyMetformin (PM) in hybrid membrane-camouflaged liposomes. Specifically, the hybrid cell membrane coating derived from the fusion of cancer cell membrane and red blood cell membrane displays excellent tumor targeting efficiency and long blood circulation property due to the innate features of both membranes. After collaboration with aPD-L1-based immune checkpoint blockade therapy, a boosted immunotherapeutic effect is obtained due to elevated dendritic cell maturation and T cell activation. Significantly, laser-irradiated BPP@RTL combined with aPD-L1 effectively eliminates primary tumors and inhibits lung metastasis in 4T1 breast tumor model, offering a promising treatment plan to develop personalized antitumor strategy.
Collapse
Affiliation(s)
- Yuqi Cao
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Lu Tang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Cong Fu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Yue Yin
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Hening Liu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Jingwen Feng
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Jifan Gao
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Weijie Shu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Zixuan Li
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Yuanbo Zhu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Wei Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing 211198, P. R. China
| |
Collapse
|
32
|
Chan PY, Corrie PG. Curing Stage IV Melanoma: Where Have We Been and Where Are We? Am Soc Clin Oncol Educ Book 2024; 44:e438654. [PMID: 38669609 DOI: 10.1200/edbk_438654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
Little more than 10 years ago, metastatic melanoma was considered to have one of the poorest cancer outcomes, associated with a median overall survival of 6-8 months. Cytotoxic chemotherapy offered modest response rates of 20%-30%, but no clear survival benefit. Patients were routinely enrolled in clinical trials as their first-line therapy in the search for effective novel therapeutics. Remarkable developments in molecular biology, cancer genomics, immunology, and drug discovery have dominated the early part of the 21st century, and nowhere have the benefits been better realized than in the transformation of outcomes for patients with metastatic melanoma: since 2011, 14 new agents have been approved that significantly increase survival, with long-term remissions and, possibly now, potential for cure. Even so, there is still much work to be done, given that most treated patients still die of their disease. Although most survival gains have so far been realized for cutaneous melanoma, improving treatment options for those 10% of patients with rarer, noncutaneous melanomas is a high priority. Key novel therapeutic approaches aimed at improving outcomes with potential for curing patients with melanoma are considered.
Collapse
Affiliation(s)
- Pui Ying Chan
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Pippa G Corrie
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| |
Collapse
|
33
|
Haanen J, Los C, Phan GQ, Betof Warner A. Adoptive Cell Therapy for Solid Tumors: Current Status in Melanoma and Next-Generation Therapies. Am Soc Clin Oncol Educ Book 2024; 44:e431608. [PMID: 38776509 DOI: 10.1200/edbk_431608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Lifileucel or TIL has recently been FDA approved for metastatic melanoma patients as first cell therapy for a solid tumor. We discuss roll-out of TIL as new SOC and other upcoming new cell therapies.
Collapse
Affiliation(s)
- John Haanen
- Division of Medical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
- Division of Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Christy Los
- Division of Medical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Giao Q Phan
- Division of Surgical Oncology, UConn Health, Neag Cancer Center, Farmington, CT
| | - Allison Betof Warner
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
34
|
Khan B, Qahwaji RM, Alfaifi MS, Mobashir M. Nivolumab and Ipilimumab Acting as Tormentors of Advanced Tumors by Unleashing Immune Cells and Associated Collateral Damage. Pharmaceutics 2024; 16:732. [PMID: 38931856 PMCID: PMC11207028 DOI: 10.3390/pharmaceutics16060732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/22/2024] [Accepted: 05/01/2024] [Indexed: 06/28/2024] Open
Abstract
Combining immune checkpoint inhibitors, specifically nivolumab (anti-PD-1) and ipilimumab (anti-CTLA-4), holds substantial promise in revolutionizing cancer treatment. This review explores the transformative impact of these combinations, emphasizing their potential for enhancing therapeutic outcomes across various cancers. Immune checkpoint proteins, such as PD1 and CTLA4, play a pivotal role in modulating immune responses. Blocking these checkpoints unleashes anticancer activity, and the synergy observed when combining multiple checkpoint inhibitors underscores their potential for enhanced efficacy. Nivolumab and ipilimumab harness the host's immune system to target cancer cells, presenting a powerful approach to prevent tumor development. Despite their efficacy, immune checkpoint inhibitors are accompanied by a distinct set of adverse effects, particularly immune-related adverse effects affecting various organs. Understanding these challenges is crucial for optimizing treatment strategies and ensuring patient well-being. Ongoing clinical trials are actively exploring the combination of checkpoint inhibitory therapies, aiming to decipher their synergistic effects and efficacy against diverse cancer types. This review discusses the mechanisms, adverse effects, and various clinical trials involving nivolumab and ipilimumab across different cancers, emphasizing their transformative impact on cancer treatment.
Collapse
Affiliation(s)
- Bushra Khan
- Department of Biosciences, Jamia Millia Islamia, New Delhi 110025, India;
| | - Rowaid M. Qahwaji
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 22233, Saudi Arabia;
- Hematology Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mashael S. Alfaifi
- Department of Epidemiology, Faculty of Public Health and Health Informatics, Umm Al-Qura University, Makkah 21955, Saudi Arabia;
| | - Mohammad Mobashir
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institute, Solnavägen 9, 171 65 Solna, Sweden
| |
Collapse
|
35
|
Wu LY, Park SH, Jakobsson H, Shackleton M, Möller A. Immune Regulation and Immune Therapy in Melanoma: Review with Emphasis on CD155 Signalling. Cancers (Basel) 2024; 16:1950. [PMID: 38893071 PMCID: PMC11171058 DOI: 10.3390/cancers16111950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 06/21/2024] Open
Abstract
Melanoma is commonly diagnosed in a younger population than most other solid malignancies and, in Australia and most of the world, is the leading cause of skin-cancer-related death. Melanoma is a cancer type with high immunogenicity; thus, immunotherapies are used as first-line treatment for advanced melanoma patients. Although immunotherapies are working well, not all the patients are benefitting from them. A lack of a comprehensive understanding of immune regulation in the melanoma tumour microenvironment is a major challenge of patient stratification. Overexpression of CD155 has been reported as a key factor in melanoma immune regulation for the development of therapy resistance. A more thorough understanding of the actions of current immunotherapy strategies, their effects on immune cell subsets, and the roles that CD155 plays are essential for a rational design of novel targets of anti-cancer immunotherapies. In this review, we comprehensively discuss current anti-melanoma immunotherapy strategies and the immune response contribution of different cell lineages, including tumour endothelial cells, myeloid-derived suppressor cells, cytotoxic T cells, cancer-associated fibroblast, and nature killer cells. Finally, we explore the impact of CD155 and its receptors DNAM-1, TIGIT, and CD96 on immune cells, especially in the context of the melanoma tumour microenvironment and anti-cancer immunotherapies.
Collapse
Affiliation(s)
- Li-Ying Wu
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4059, Australia;
- JC STEM Lab, Department of Otorhinolaryngology, Chinese University of Hong Kong, Shatin, Hong Kong SAR, China;
- Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Su-Ho Park
- JC STEM Lab, Department of Otorhinolaryngology, Chinese University of Hong Kong, Shatin, Hong Kong SAR, China;
- Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Haakan Jakobsson
- Department of Medical Oncology, Paula Fox Melanoma and Cancer Centre, Alfred Health, Melbourne, VIC 3004, Australia;
| | - Mark Shackleton
- Department of Medical Oncology, Paula Fox Melanoma and Cancer Centre, Alfred Health, Melbourne, VIC 3004, Australia;
- School of Translational Medicine, Monash University, Melbourne, VIC 3004, Australia
| | - Andreas Möller
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4059, Australia;
- JC STEM Lab, Department of Otorhinolaryngology, Chinese University of Hong Kong, Shatin, Hong Kong SAR, China;
- Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
36
|
Ren Z, Yang K, Zhu L, Yin D, Zhou Y. Regulatory T cells as crucial trigger and potential target for hyperprogressive disease subsequent to PD-1/PD-L1 blockade for cancer treatment. Int Immunopharmacol 2024; 132:111934. [PMID: 38574701 DOI: 10.1016/j.intimp.2024.111934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/21/2024] [Accepted: 03/24/2024] [Indexed: 04/06/2024]
Abstract
PD-1/PD-L1 blockade therapy has brought great success to cancer treatment. Nevertheless, limited beneficiary populations and even hyperprogressive disease (HPD) greatly constrain the application of PD-1/PD-L1 inhibitors in clinical treatment. HPD is a special pattern of disease progression with rapid tumor growth and even serious consequences of patient death, which requires urgent attention. Among the many predisposing causes of HPD, regulatory T cells (Tregs) are suspected because they are amplified in cases of HPD. Tregs express PD-1 thus PD-1/PD-L1 blockade therapy may have an impact on Tregs which leads to HPD. Tregs are a subset of CD4+ T cells expressing FoxP3 and play critical roles in suppressing immunity. Tregs migrate toward tumors in the presence of chemokines to suppress antitumor immune responses, causing cancer cells to grow and proliferate. Studies have shown that deleting Tregs could enhance the efficacy of PD-1/PD-L1 blockade therapy and reduce the occurrence of HPD. This suggests that immunotherapy combined with Treg depletion may be an effective means of avoiding HPD. In this review, we summarized the immunosuppressive-related functions of Tregs in antitumor therapy and focused on advances in therapy combining Tregs depletion with PD-1/PD-L1 blockade in clinical studies. Moreover, we provided an outlook on Treg-targeted HPD early warning for PD-1/PD-L1 blockade therapy.
Collapse
Affiliation(s)
- Zhe Ren
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450000, Henan, China
| | - Kaiqing Yang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Lin Zhu
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Detao Yin
- Department of Thyroid Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China.
| | - Yubing Zhou
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China.
| |
Collapse
|
37
|
Holmstroem RB, Pedersen S, Jurlander R, Madsen K, Donia M, Ruhlmann CH, Schmidt H, Haslund CA, Bastholt L, Svane IM, Ellebaek E. Outcome of adjuvant immunotherapy in a real-world nation-wide cohort of patients with melanoma. Eur J Cancer 2024; 202:114023. [PMID: 38518533 DOI: 10.1016/j.ejca.2024.114023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/04/2024] [Accepted: 03/12/2024] [Indexed: 03/24/2024]
Abstract
BACKGROUND Clinical trials have demonstrated promising outcomes for adjuvant immunotherapy in patients with resected melanoma. Real-life data provide valuable insights to support patient guidance and treatment decisions. METHODS Observational population-based study examining a national cohort of patients with resected stage III-IV melanoma referred for adjuvant therapy. Data were extracted from the Danish Metastatic Melanoma Database (DAMMED). RESULTS Between November 2018 and January 2022, 785 patients received adjuvant anti-PD-1. The majority had stage III resected melanoma (87%), normal LDH levels (80%), and performance score 0 (87%). Patients were followed for a median of 25.6 months (95%CI 24-28). The median recurrence-free survival (RFS) and melanoma-specific survival (MSS) were not reached. The RFS was 78% (95%CI 75-81), 66% (63-70), and 59% (55-63); MSS was 97% (95-98), 93% (91-95), and 87% (84-90) at 1-, 2-, and 3-year; respectively. Less than half (42%) of the patients finalized planned therapy, 32% discontinued due to toxicity, and 19% due to melanoma recurrence. Patients discontinuing adjuvant treatment prematurely, without recurrence, had similar outcomes as patients finalizing therapy. In a multivariable analysis, ipilimumab plus nivolumab did not improve outcomes compared to ipilimumab monotherapy as a first-line metastatic treatment after adjuvant anti-PD-1. CONCLUSION Survival outcomes in real-world patients with melanoma treated with adjuvant anti-PD-1 align with results from the randomized controlled trials. Patients discontinuing therapy prematurely, for other reasons than recurrence, had similar outcomes as patients finalizing planned treatment. First-line metastatic treatment with ipilimumab and nivolumab post-adjuvant anti-PD-1 did not show improved outcomes compared to ipilimumab/anti-PD-1 monotherapy.
Collapse
Affiliation(s)
- Rikke B Holmstroem
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Denmark
| | - Sidsel Pedersen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Denmark
| | - Rebecca Jurlander
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Denmark
| | - Kasper Madsen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Denmark
| | - Marco Donia
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Denmark
| | | | - Henrik Schmidt
- Department of Oncology, Aarhus University Hospital, Denmark
| | | | - Lars Bastholt
- Department of Oncology, Odense University Hospital, Denmark
| | - Inge Marie Svane
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Denmark.
| | - Eva Ellebaek
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Denmark.
| |
Collapse
|
38
|
Rosu OA, Tolea MI, Parosanu AI, Stanciu MI, Cotan HT, Nitipir C. Challenges in the Diagnosis and Treatment of Oral Amelanotic Malignant Melanoma: A Case Report. Cureus 2024; 16:e57875. [PMID: 38725781 PMCID: PMC11079336 DOI: 10.7759/cureus.57875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/08/2024] [Indexed: 05/12/2024] Open
Abstract
Oral malignant melanoma (OMM) is extremely rare and usually has a poor prognosis. Early diagnosis is very important and can improve survival but it is usually difficult due to a lack of symptomatology. We present the first case of a 39-year-old East European woman with oral amelanotic melanoma, who underwent surgery and adjuvant immunotherapy; however, after six months, she developed local recurrence. The patient continued immunotherapy with external radiotherapy targeting the oral tumor recurrence. During therapy, imagistic reevaluation brought evidence of bones, lungs, liver, endotracheal, and brain metastases. Histological differential diagnosis between amelanotic OMM and leiomyosarcoma was necessary to establish the right course of treatment. A series of complications further delayed chemotherapy administration, making the treatment in this case very challenging. The patient had a significant, although late response to immunotherapy, and maintained a good performance status during disease progression with a survival of 15 months until present.
Collapse
Affiliation(s)
- Oana A Rosu
- Oncology, Elias Emergency University Hospital, Bucuresti, ROU
| | | | - Andreea I Parosanu
- Oncology, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
- Oncology, Elias Emergency University Hospital, Bucharest, ROU
| | - Miruna I Stanciu
- Medical Oncology, Elias Emergency University Hospital, Bucharest, ROU
| | - Horia T Cotan
- Oncology, Elias Emergency University Hospital, Bucharest, ROU
| | | |
Collapse
|
39
|
Boutros A, Croce E, Ferrari M, Gili R, Massaro G, Marconcini R, Arecco L, Tanda ET, Spagnolo F. The treatment of advanced melanoma: Current approaches and new challenges. Crit Rev Oncol Hematol 2024; 196:104276. [PMID: 38295889 DOI: 10.1016/j.critrevonc.2024.104276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 01/15/2024] [Accepted: 01/26/2024] [Indexed: 02/17/2024] Open
Abstract
In recent years, advances in melanoma treatment have renewed patient hope. This comprehensive review emphasizes the evolving treatment landscape, particularly highlighting first-line strategies and the interplay between immune-checkpoint inhibitors (ICIs) and targeted therapies. Ipilimumab plus nivolumab has achieved the best median overall survival, exceeding 70 months. However, the introduction of new ICIs, like relatlimab, has added complexity to first-line therapy decisions. Our aim is to guide clinicians in making personalized treatment decisions. Various features, including brain metastases, PD-L1 expression, BRAF mutation, performance status, and prior adjuvant therapy, significantly impact the direction of advanced melanoma treatment. We also provide the latest insights into the treatment of rare melanoma subtypes, such as uveal melanoma, where tebentafusp has shown promising improvements in overall survival for metastatic uveal melanoma patients. This review provides invaluable insights for clinicians, enabling informed treatment choices and deepening our understanding of the multifaceted challenges associated with advanced melanoma management.
Collapse
Affiliation(s)
- Andrea Boutros
- Skin Cancer Unit, U.O. Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, Genova, Italy; Department of Internal Medicine and Medical Sciences (DiMI), School of Medicine, University of Genova, Genova, Italy.
| | - Elena Croce
- Skin Cancer Unit, U.O. Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Marco Ferrari
- Azienda Ospedaliero Universitaria Pisana, Medical Oncology Unit, Pisa, Italy
| | - Riccardo Gili
- Skin Cancer Unit, U.O. Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, Genova, Italy; Department of Internal Medicine and Medical Sciences (DiMI), School of Medicine, University of Genova, Genova, Italy
| | - Giulia Massaro
- Unit of Medical Oncology, Careggi University-Hospital, 50134 Florence, Italy
| | - Riccardo Marconcini
- Azienda Ospedaliero Universitaria Pisana, Medical Oncology Unit, Pisa, Italy
| | - Luca Arecco
- Department of Internal Medicine and Medical Sciences (DiMI), School of Medicine, University of Genova, Genova, Italy; Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Enrica Teresa Tanda
- Skin Cancer Unit, U.O. Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Francesco Spagnolo
- Skin Cancer Unit, U.O. Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, Genova, Italy; Department of Surgical Sciences and Integrated Diagnostics (DISC), Plastic Surgery Division, University of Genova, Genova, Italy
| |
Collapse
|
40
|
Wisdom AJ, Barker CA, Chang JY, Demaria S, Formenti S, Grassberger C, Gregucci F, Hoppe BS, Kirsch DG, Marciscano AE, Mayadev J, Mouw KW, Palta M, Wu CC, Jabbour SK, Schoenfeld JD. The Next Chapter in Immunotherapy and Radiation Combination Therapy: Cancer-Specific Perspectives. Int J Radiat Oncol Biol Phys 2024; 118:1404-1421. [PMID: 38184173 DOI: 10.1016/j.ijrobp.2023.12.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 12/20/2023] [Accepted: 12/30/2023] [Indexed: 01/08/2024]
Abstract
Immunotherapeutic agents have revolutionized cancer treatment over the past decade. However, most patients fail to respond to immunotherapy alone. A growing body of preclinical studies highlights the potential for synergy between radiation therapy and immunotherapy, but the outcomes of clinical studies have been mixed. This review summarizes the current state of immunotherapy and radiation combination therapy across cancers, highlighting existing challenges and promising areas for future investigation.
Collapse
Affiliation(s)
- Amy J Wisdom
- Harvard Radiation Oncology Program, Boston, Massachusetts
| | - Christopher A Barker
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Joe Y Chang
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medicine, New York, New York
| | - Silvia Formenti
- Department of Radiation Oncology, Weill Cornell Medicine, New York, New York
| | - Clemens Grassberger
- Department of Radiation Oncology, University of Washington, Fred Hutch Cancer Center, Seattle, Washington
| | - Fabiana Gregucci
- Department of Radiation Oncology, Weill Cornell Medicine, New York, New York
| | - Bradford S Hoppe
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, Florida
| | - David G Kirsch
- Department of Radiation Oncology, University of Toronto, Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Ariel E Marciscano
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Jyoti Mayadev
- Department of Radiation Oncology, UC San Diego School of Medicine, San Diego, California
| | - Kent W Mouw
- Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Manisha Palta
- Department of Radiation Oncology, Duke Cancer Center, Durham, North Carolina
| | - Cheng-Chia Wu
- Department of Radiation Oncology, Columbia University Irving Medical Center, New York, New York
| | - Salma K Jabbour
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey.
| | - Jonathan D Schoenfeld
- Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Institute, Boston, Massachusetts.
| |
Collapse
|
41
|
Liu C, Cheng X, Han K, Hong L, Hao S, Sun X, Xu J, Li B, Jin D, Tian W, Jin Y, Wang Y, Fang W, Bao X, Zhao P, Chen D. A novel molecular subtyping based on multi-omics analysis for prognosis predicting in colorectal melanoma: A 16-year prospective multicentric study. Cancer Lett 2024; 585:216663. [PMID: 38246221 DOI: 10.1016/j.canlet.2024.216663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/13/2024] [Accepted: 01/18/2024] [Indexed: 01/23/2024]
Abstract
Colorectal melanoma (CRM) is a rare malignant tumor with severe complications, and there is currently a lack of systematic research. We conducted a study that combined proteomics and mutation data of CRM from a cohort of three centers over a 16-years period (2005-2021). The patients were divided into a training set consisting of two centers and a testing set comprising the other center. Unsupervised clustering was conducted on the training set to form two molecular subtypes for clinical characterization and functional analysis. The testing set was used to validate the survival differences between the two subtypes. The comprehensive analysis identified two subtypes of CRM: immune exhausted C1 cluster and DNA repair C2 cluster. The former subtype exhibited characteristics of metabolic disturbance, immune suppression, and poor prognosis, along with APC mutations. A machine learning algorithm named Support Vector Machine (SVM) was applied to predict the classification of CRM patients based on protein expression in the external testing cohort. Two subtypes of primary CRM with clinical and proteomic characteristics provides a reference for subsequent diagnosis and treatments.
Collapse
Affiliation(s)
- Chuan Liu
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, People's Republic of China
| | - Xiaofei Cheng
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, People's Republic of China
| | - Kai Han
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Libing Hong
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, People's Republic of China; The Second Clinical School, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Shuqiang Hao
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, People's Republic of China
| | - Xuqi Sun
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, People's Republic of China
| | - Jingfeng Xu
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, People's Republic of China
| | - Benfeng Li
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, People's Republic of China
| | - Dongqing Jin
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, People's Republic of China
| | - Weihong Tian
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, 212013, People's Republic of China
| | - Yuzhi Jin
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, People's Republic of China
| | - Yanli Wang
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, People's Republic of China
| | - Weijia Fang
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, People's Republic of China
| | - Xuanwen Bao
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, People's Republic of China.
| | - Peng Zhao
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, People's Republic of China.
| | - Dong Chen
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, People's Republic of China.
| |
Collapse
|
42
|
Revach OY, Cicerchia AM, Shorer O, Petrova B, Anderson S, Park J, Chen L, Mehta A, Wright SJ, McNamee N, Tal-Mason A, Cattaneo G, Tiwari P, Xie H, Sweere JM, Cheng LC, Sigal N, Enrico E, Miljkovic M, Evans SA, Nguyen N, Whidden ME, Srinivasan R, Spitzer MH, Sun Y, Sharova T, Lawless AR, Michaud WA, Rasmussen MQ, Fang J, Palin CA, Chen F, Wang X, Ferrone CR, Lawrence DP, Sullivan RJ, Liu D, Sachdeva UM, Sen DR, Flaherty KT, Manguso RT, Bod L, Kellis M, Boland GM, Yizhak K, Yang J, Kanarek N, Sade-Feldman M, Hacohen N, Jenkins RW. Disrupting CD38-driven T cell dysfunction restores sensitivity to cancer immunotherapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.12.579184. [PMID: 38405985 PMCID: PMC10888727 DOI: 10.1101/2024.02.12.579184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
A central problem in cancer immunotherapy with immune checkpoint blockade (ICB) is the development of resistance, which affects 50% of patients with metastatic melanoma1,2. T cell exhaustion, resulting from chronic antigen exposure in the tumour microenvironment, is a major driver of ICB resistance3. Here, we show that CD38, an ecto-enzyme involved in nicotinamide adenine dinucleotide (NAD+) catabolism, is highly expressed in exhausted CD8+ T cells in melanoma and is associated with ICB resistance. Tumour-derived CD38hiCD8+ T cells are dysfunctional, characterised by impaired proliferative capacity, effector function, and dysregulated mitochondrial bioenergetics. Genetic and pharmacological blockade of CD38 in murine and patient-derived organotypic tumour models (MDOTS/PDOTS) enhanced tumour immunity and overcame ICB resistance. Mechanistically, disrupting CD38 activity in T cells restored cellular NAD+ pools, improved mitochondrial function, increased proliferation, augmented effector function, and restored ICB sensitivity. Taken together, these data demonstrate a role for the CD38-NAD+ axis in promoting T cell exhaustion and ICB resistance, and establish the efficacy of CD38 directed therapeutic strategies to overcome ICB resistance using clinically relevant, patient-derived 3D tumour models.
Collapse
Affiliation(s)
- Or-Yam Revach
- Mass General Cancer Center, Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Angelina M. Cicerchia
- Mass General Cancer Center, Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Ofir Shorer
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Boryana Petrova
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Boston Children’s Hospital, Boston, MA, USA
| | - Seth Anderson
- Mass General Cancer Center, Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Joshua Park
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Lee Chen
- Computer Science and Artificial Intelligence Lab, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Arnav Mehta
- Mass General Cancer Center, Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Niamh McNamee
- Harvard Medical School, Boston, MA, USA
- Division of Thoracic Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Aya Tal-Mason
- Harvard Medical School, Boston, MA, USA
- Division of Thoracic Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Giulia Cattaneo
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Payal Tiwari
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Hongyan Xie
- Mass General Cancer Center, Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | | | | | | | | | | | | | | | | | - Matthew H. Spitzer
- Teiko Bio, Salt Lake City, UT, USA
- Department of Otolaryngology-Head and Neck Cancer, University of California, San Francisco, San Francisco, CA, USA
- Department of Microbiology & Immunology, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158; Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Yi Sun
- Mass General Cancer Center, Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Tatyana Sharova
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Aleigha R. Lawless
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - William A. Michaud
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Martin Q. Rasmussen
- Mass General Cancer Center, Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jacy Fang
- Mass General Cancer Center, Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Claire A. Palin
- Mass General Cancer Center, Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Feng Chen
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Xinhui Wang
- Harvard Medical School, Boston, MA, USA
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Cristina R. Ferrone
- Harvard Medical School, Boston, MA, USA
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
- Department of Surgery, Cedars-Sinai Medical Center Los Angeles, CA, USA
| | - Donald P. Lawrence
- Mass General Cancer Center, Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Ryan J. Sullivan
- Mass General Cancer Center, Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - David Liu
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Uma M. Sachdeva
- Harvard Medical School, Boston, MA, USA
- Division of Thoracic Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Debattama R. Sen
- Mass General Cancer Center, Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Keith T. Flaherty
- Mass General Cancer Center, Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Robert T. Manguso
- Mass General Cancer Center, Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Lloyd Bod
- Mass General Cancer Center, Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Manolis Kellis
- Department of Pathology, Boston Children’s Hospital, Boston, MA, USA
| | - Genevieve M. Boland
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Keren Yizhak
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Jiekun Yang
- Computer Science and Artificial Intelligence Lab, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Naama Kanarek
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Pathology, Boston Children’s Hospital, Boston, MA, USA
| | - Moshe Sade-Feldman
- Mass General Cancer Center, Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Nir Hacohen
- Mass General Cancer Center, Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Russell W. Jenkins
- Mass General Cancer Center, Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
43
|
Fletcher KA, Johnson DB. Investigational Approaches for Treatment of Melanoma Patients Progressing After Standard of Care. Cancer J 2024; 30:126-131. [PMID: 38527267 DOI: 10.1097/ppo.0000000000000702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
ABSTRACT The advent of effective immunotherapy, specifically cytotoxic T-lymphocyte associated protein 4 and programmed cell death 1 inhibitors, as well as targeted therapy including BRAF/MEK inhibitors, has dramatically changed the prognosis for metastatic melanoma patients. Up to 50% of patients may experience long-term survival currently. Despite these advances in melanoma treatment, many patients still progress and die of their disease. As such, there are many studies aimed at providing new treatment options for this population. Therapies currently under investigation include, but are not limited to, novel immunotherapies, targeted therapies, tumor-infiltrating lymphocytes and other cellular therapies, oncolytic viral therapy and other injectables, and fecal microbiota transplant. In this review, we discuss the emerging treatment options for metastatic melanoma patients who have progressed on standard of care treatments.
Collapse
Affiliation(s)
| | - Douglas B Johnson
- Department of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
44
|
Los C, Klobuch S, Haanen JBAG. Tumor-Infiltrating Lymphocyte and Other Cell Therapies for Metastatic Melanoma. Cancer J 2024; 30:113-119. [PMID: 38527265 DOI: 10.1097/ppo.0000000000000705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
ABSTRACT Major progress in prolonging survival of patients with advanced melanoma has been made in the past decade because of the development and approval of immune checkpoint inhibitor and targeted therapies. However, for nonresponding or relapsing patients, their prognosis is still dismal. Based on clinical trial data, treatment with adoptive cell therapies holds great promise. In patients with metastatic melanoma progressing on or nonresponsive to single-agent anti-programmed cell death 1, infusion of tumor-infiltrating lymphocytes can produce responses in up to half of patients, with durable complete responses in up to 20%. Genetic modification of peripheral blood T cells with T-cell receptors derived from tumor-specific T cells, or with chimeric antigen receptors, has the potential to further improve treatment outcomes in this refractory population. In this review, we will discuss the historical development, current status, and future perspectives of adoptive T-cell therapies in melanoma.
Collapse
Affiliation(s)
- Christy Los
- From the Division of Molecular Oncology and Immunology, Oncode Institute, Netherlands Cancer Institute
| | - Sebastian Klobuch
- Department of Medical Oncology, Antoni van Leeuwenhoek/Netherlands Cancer Institute, Amsterdam
| | | |
Collapse
|
45
|
Namikawa K, Nakano E, Ogata D, Yamazaki N. Long-term survival with systemic therapy in the last decade: Can melanoma be cured? J Dermatol 2024; 51:343-352. [PMID: 38358050 PMCID: PMC11484129 DOI: 10.1111/1346-8138.17147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 02/16/2024]
Abstract
Immune checkpoint inhibitors have been shown to prolong survival of patients with several types of cancer, and the finding was first established in melanoma. Previously, systemic therapy for advanced melanoma aimed only at tumor control and palliation of symptoms. However, in recent years, some patients who received systemic therapy have achieved a complete response and survived without continuous treatment for more than several years. This review discusses the long-term survival rates achieved with currently used systemic therapies and their future perspectives. Long-term survival is currently most likely to be achieved with the use of the standard-dose combination of nivolumab plus ipilimumab, however, this regimen is associated with a high frequency of serious or persistent immune-related adverse events. Several new anti-PD-1-based combination therapies with a better risk-benefit balance are currently under development. Although the acral and mucosal subtypes tend to be less responsive to immune checkpoint inhibitors, anti-PD-1-based combination therapy should continue to be investigated for these subtypes owing to its potential for better long-term survival. With the development of efficacious immunotherapy and targeted therapy, it is important to determine the optimal duration of systemic therapy to avoid unnecessary health and financial burdens as well as to improve efforts to support long-term cancer survivors. As the goal of systemic therapy shifts from tumor control to long-term survival, in future clinical trials, long-term clinical outcomes should be evaluated to assess the benefits of novel agents.
Collapse
Affiliation(s)
- Kenjiro Namikawa
- Department of Dermatologic OncologyNational Cancer Center HospitalTokyoJapan
| | - Eiji Nakano
- Department of Dermatologic OncologyNational Cancer Center HospitalTokyoJapan
| | - Dai Ogata
- Department of Dermatologic OncologyNational Cancer Center HospitalTokyoJapan
| | - Naoya Yamazaki
- Department of Dermatologic OncologyNational Cancer Center HospitalTokyoJapan
| |
Collapse
|
46
|
Di Giacomo AM, Lahn M, Eggermont AM, Fox B, Ibrahim R, Sharma P, Allison JP, Maio M. The future of targeting cytotoxic T-lymphocyte-associated protein-4: Is there a role? Eur J Cancer 2024; 198:113501. [PMID: 38169219 DOI: 10.1016/j.ejca.2023.113501] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 12/13/2023] [Indexed: 01/05/2024]
Abstract
The 2022 yearly Think Tank Meeting in Siena, Tuscany (Italy), organized by the Italian Network for Tumor Biotherapy (NIBIT) Foundation, the Parker Institute for Cancer Immunotherapy and the World Immunotherapy Council, included a focus on the future of integrating and expanding the use of targeting cytotoxic T-lymphocyte-associated protein 4 (CTLA-4). The conference members exchanged their views on the lessons from targeting CTLA-4 and compared the effect to the impact of blocking Programmed cell death protein 1 (PD1) or its ligand (PDL1). The increasing experience with both therapeutic approaches and their combination suggests that targeting CTLA-4 may lead to more durable responses for a sizeable proportion of patients, though the specific mechanism is not entirely understood. Overcoming toxicity of blocking CTLA-4 is currently being addressed with different doses and dose regimens, especially when combined with PD1/PDL1 blocking antibodies. Novel therapeutics targeting CTLA-4 hold the promise to reduce toxicities and thus allow different combination strategies in the future. On the whole, the consent was that targeting CTLA-4 remains an important strategy to improve the efficacy of cancer immunotherapies.
Collapse
Affiliation(s)
- Anna Maria Di Giacomo
- University of Siena, Siena, Italy; Center for Immuno-Oncology. University Hospital of Siena, Viale Bracci, 16, Siena, Italy; NIBIT Foundation Onlus, Italy
| | - Michael Lahn
- IOnctura SA, Avenue Secheron 15, Geneva, Switzerland
| | - Alexander Mm Eggermont
- Princess Máxima Center and the University Medical Center Utrecht, Heidelberglaan 25, 3584 Utrecht, the Netherlands; Comprehensive Cancer Center Munich of the Technical University Munich and the Ludwig Maximiliaan University, Munich, Germany
| | - Bernard Fox
- Earle A. Chiles Research Institute at the Robert W. Franz Cancer Center, 4805 NE Glisan St. Suite 2N35 Portland, OR 97213, USA
| | - Ramy Ibrahim
- Parker Institute for Cancer Immunotherapy, 1 Letterman Drive, D3500, San Francisco, CA, USA
| | - Padmanee Sharma
- Department of Genitourinary Medical Oncology, Division of Cancer Medicine, MD Anderson, 1515 Holcombe Blvd, Houston, Texas 77030, USA
| | - James P Allison
- James P Allison Institute, MD Anderson, 1515 Holcombe Blvd, Texas 77030, USA
| | - Michele Maio
- University of Siena, Siena, Italy; Center for Immuno-Oncology. University Hospital of Siena, Viale Bracci, 16, Siena, Italy; NIBIT Foundation Onlus, Italy.
| |
Collapse
|
47
|
Sondak VK, Atkins MB, Messersmith H, Provenzano A, Seth R, Agarwala SS. Systemic Therapy for Melanoma: ASCO Guideline Update Q and A. JCO Oncol Pract 2024; 20:173-177. [PMID: 38039436 DOI: 10.1200/op.23.00675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 11/01/2023] [Indexed: 12/03/2023] Open
Abstract
This Q&A answers questions regarding ASCO's recent Systemic Therapy for Melanoma guideline.
Collapse
Affiliation(s)
- Vernon K Sondak
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | | | | | | | - Rahul Seth
- SUNY Upstate Medical University, Syracuse, NY
| | - Sanjiv S Agarwala
- Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| |
Collapse
|
48
|
Lamarca A, Moreno V, Gambardella V, Cervantes A. In the literature: September 2023. ESMO Open 2023; 8:102032. [PMID: 37852035 PMCID: PMC10590842 DOI: 10.1016/j.esmoop.2023.102032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 09/12/2023] [Indexed: 10/20/2023] Open
Affiliation(s)
- A Lamarca
- Department of Medical Oncology - Oncohealth Institute, Fundación Jiménez Díaz University Hospital, Madrid, Spain; Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - V Moreno
- START-FJD Phase I Unit, Department of Medical Oncology, Fundación Jiménez Díaz University Hospital, Madrid
| | - V Gambardella
- INCLIVA Biomedical Research Institute, Hospital Clínico Universitario, Universidad de Valencia, Valencia; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - A Cervantes
- INCLIVA Biomedical Research Institute, Hospital Clínico Universitario, Universidad de Valencia, Valencia; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
49
|
Zhang P, Wang Y, Miao Q, Chen Y. The therapeutic potential of PD-1/PD-L1 pathway on immune-related diseases: Based on the innate and adaptive immune components. Biomed Pharmacother 2023; 167:115569. [PMID: 37769390 DOI: 10.1016/j.biopha.2023.115569] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 09/30/2023] Open
Abstract
Currently, immunotherapy targeting programmed cell death 1 (PD-1) or programmed death ligand 1 (PD-L1) has revolutionized the treatment strategy of human cancer patients. Meanwhile, PD-1/PD-L1 pathway has also been implicated in the pathogenesis of many immune-related diseases, such as autoimmune diseases, chronic infection diseases and adverse pregnancy outcomes, by regulating components of the innate and adaptive immune systems. Given the power of the new therapy, a better understanding of the regulatory effects of PD-1/PD-L1 pathway on innate and adaptive immune responses in immune-related diseases will facilitate the discovery of novel biomarkers and therapeutic drug targets. Targeting this pathway may successfully halt or potentially even reverse these pathological processes. In this review, we discuss recent major advances in PD-1/PD-L1 axis regulating innate and adaptive immune components in immune-related diseases. We reveal that the impact of PD-1/PD-L1 axis on the immune system is complex and manifold and multi-strategies on the targeted PD-1/PD-L1 axis are taken in the treatment of immune-related diseases. Consequently, targeting PD-1/PD-L1 pathway, alone or in combination with other treatments, may represent a novel strategy for future therapeutic intervention on immune-related diseases.
Collapse
Affiliation(s)
- Peng Zhang
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention (China Medical University), Ministry of Education, Shenyang 110122, Liaoning, China; Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang 110122, Liaoning, China
| | - Yuting Wang
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention (China Medical University), Ministry of Education, Shenyang 110122, Liaoning, China; Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang 110122, Liaoning, China
| | - Qianru Miao
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention (China Medical University), Ministry of Education, Shenyang 110122, Liaoning, China; Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang 110122, Liaoning, China
| | - Ying Chen
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention (China Medical University), Ministry of Education, Shenyang 110122, Liaoning, China; Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang 110122, Liaoning, China.
| |
Collapse
|