201
|
Mitamura A, Tsujinaka S, Nakano T, Sawada K, Shibata C. Treatment Strategies for Locoregional Recurrence in Esophageal Squamous-Cell Carcinoma: An Updated Review. Cancers (Basel) 2024; 16:2539. [PMID: 39061179 PMCID: PMC11274925 DOI: 10.3390/cancers16142539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/02/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Emerging evidence has shown remarkable advances in the multimodal treatment of esophageal squamous-cell carcinoma. Despite these advances, the oncological outcomes for advanced esophageal cancer remain controversial due to the frequent observation of local recurrence in the regional or other lymph nodes and distant metastasis after curative treatment. For cases of locoregional recurrence in the cervical lymph nodes alone, salvage surgery with lymph node dissection generally provides a good prognosis. However, if recurrence occurs in multiple regions, the oncological efficacy of surgery may be limited. Radiotherapy/chemoradiotherapy can be employed for unresectable or recurrent cases, as well as for selected cases in neo- or adjuvant settings. Dose escalation and toxicity are potential issues with conventional three-dimensional conformal radiotherapy; however, more precise therapeutic efficacy can be obtained using technical modifications with improved targeting and conformality, or with the use of proton beam therapy. The introduction of immune checkpoint inhibitors, including pembrolizumab or nivolumab, in addition to chemotherapy, has been shown to improve the overall survival in unresectable, advanced/recurrent cases. For patients with lymph node recurrence in multiple regions, chemotherapy (5-fluorouracil [5-FU] plus cisplatin) and combination therapy with nivolumab and ipilimumab have shown comparable oncological efficacy. Further prospective studies are needed to improve the treatment outcomes in patients with esophageal cancer with locoregional recurrence.
Collapse
Affiliation(s)
| | - Shingo Tsujinaka
- Division of Gastroenterological Surgery, Department of Surgery, Tohoku Medical and Pharmaceutical University, Sendai 983-8536, Japan
| | | | | | | |
Collapse
|
202
|
Younis A, Gribben J. Immune Checkpoint Inhibitors: Fundamental Mechanisms, Current Status and Future Directions. IMMUNO 2024; 4:186-210. [DOI: 10.3390/immuno4030013] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
Immune checkpoint inhibitors (ICI) are a promising form of immunotherapy that have significantly changed the therapeutic landscape for many advanced cancers. They have shown unique clinical benefit against a broad range of tumour types and a strong overall impact on survival in studied patient populations. However, there are still many limitations holding back this immunotherapy from reaching its full potential as a possible curative option for advanced cancer patients. A great deal of research is being undertaken in the hope of driving advancements in this area, building a better understanding of the mechanisms behind immune checkpoint inhibition and ultimately developing more effective, safer, and wider-reaching agents. Taking into account the current literature on this topic, this review aims to explore in depth the basis of the use of ICIs in the treatment of advanced cancers, evaluate its efficacy and safety, consider its current limitations, and finally reflect on what the future holds for this very promising form of cancer immunotherapy.
Collapse
Affiliation(s)
- Abdullah Younis
- Barts and the London School of Medicine and Dentistry, London E1 2AD, UK
| | - John Gribben
- Barts Cancer Institute, Queen Mary University of London, London EC1M 6AU, UK
| |
Collapse
|
203
|
Burri E, Mangana J, Cheng PF, Schneider A, Weber A, Dummer R, Ramelyte E. Infliximab in steroid-refractory immune-related hepatitis does not demonstrate hepatotoxicity and may shorten time on steroids. J Immunother Cancer 2024; 12:e008074. [PMID: 38969522 PMCID: PMC11227818 DOI: 10.1136/jitc-2023-008074] [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: 05/02/2024] [Indexed: 07/07/2024] Open
Abstract
BACKGROUND Immune-related hepatitis (irHepatitis) is a relatively common immune-related adverse event (irAE) of checkpoint inhibitors. Often, it responds well to steroids; however, in refractory cases, further therapy is needed. Anti-tumor necrosis factor (TNF) antibodies are used for management of multiple irAEs, but there are little data in irHepatitis. Here, we report on safety and efficacy of infliximab in 10 cases of steroid-refractory irHepatitis. METHODS We retrospectively reviewed patients treated with infliximab for steroid-refractory grade ≥3 irHepatitis at the Department of Dermatology, University Hospital Zurich. The positive response to infliximab was defined as no further increase in alanine aminotransferase (ALT)/aspartate aminotransferase (AST) above 50% than at the time of first infliximab infusion and control of irHepatitis without therapies other than steroids and infliximab. RESULTS 10 patients with steroid-resistant irHepatitis grade ≥3 were treated with infliximab 5 mg/kg, of whom 7 (70%) responded positively. In two cases, the liver values increased over 50% before the irHepatitis could be controlled. In another case, therapies other than infliximab and steroids were given. At the median follow-up of 487 days, 90% of the patients demonstrated resolved irHepatitis without AST/ALT elevation following infliximab infusions. CONCLUSIONS Treatment of irHepatitis with infliximab did not result in hepatotoxicity and led to long-lasting positive response in 9 of 10 of the cases. Further research is needed to evaluate the role of anti-TNF antibodies in management of irHepatitis.
Collapse
Affiliation(s)
- Elias Burri
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland
- Medical Faculty, University of Zurich, Zurich, Switzerland
| | - Joanna Mangana
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland
- Medical Faculty, University of Zurich, Zurich, Switzerland
| | - Phil F Cheng
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland
- Medical Faculty, University of Zurich, Zurich, Switzerland
| | - Alia Schneider
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Achim Weber
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Reinhard Dummer
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland
- Medical Faculty, University of Zurich, Zurich, Switzerland
| | - Egle Ramelyte
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland
- Medical Faculty, University of Zurich, Zurich, Switzerland
| |
Collapse
|
204
|
Tian Y, Yin Z, Zhang C, Li Z, Wang Y, Zhang K, Chen F, Dang Q. Differences in the risk of immune-related pneumonitis between PD-1 and PD-L1 inhibitors: a meta-analysis according to the new mirror-principle and PRISMA guidelines. Cancer Immunol Immunother 2024; 73:162. [PMID: 38953977 PMCID: PMC11219650 DOI: 10.1007/s00262-024-03736-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 05/15/2024] [Indexed: 07/04/2024]
Abstract
PURPOSE To compare the risk of immune-associated pneumonitis between PD-1 and PD-L1 inhibitors, the meta-analysis was designed. METHOD The difference in risk of immune-associated pneumonitis between PD-1 and PD-L1 inhibitors was assessed by two different meta-analysis methods, the Mirror-pairing and the PRISMA guidelines. RESULTS A total of eighty-eight reports were used for meta-analysis, while thirty-two studies were used for the Mirror-pairing. Both PD-1 and PD-L1 inhibitors (used alone or combined with chemotherapy) increased the risk of developing immune-related pneumonitis (P < 0.00001; P < 0.00001). Based on indirect analyses results (subgroup analyses), the risk of PD-L1-induced pneumonitis was weaker than that of PD-1 inhibitors when the control group was chemotherapy (OR = 3.33 vs. 5.43) or placebo (OR = 2.53 vs. 3.19), while no obvious significant differences were found (P = 0.17; P = 0.53). For the Mirror-pairing-based meta-analysis, the risk of PD-1-induced pneumonitis was significantly higher than that of PD-L1 inhibitors (OR = 1.46, 95%CI [1.08, 1.98], I2 = 0%, Z = 2.47 (P = 0.01)). However, this difference was not significant, when they were combined with chemotherapy (OR = 1.05, 95%CI [0.68, 1.60], I2 = 38%, Z = 0.21 (P = 0.84)). CONCLUSION Both PD-1 and PD-L1 inhibitors increased the risk of immune-related pneumonitis, while the risk of PD-1-induced pneumonitis was significantly higher than that of PD-L1 inhibitors.
Collapse
Affiliation(s)
- Yuan Tian
- Phase I Clinical Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University, and Shandong Academy of Medical Sciences, No. 440, Jiyan Road, Huaiyin District, Jinan City, 250117, Shandong, People's Republic of China
- Radiotherapy Department, Shandong Second Provincial General Hospital, Shandong University, Jinan, 250299, Shandong, People's Republic of China
| | - Zongxiu Yin
- Department of Pulmonary and Critical Care Medicine, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, Shandong, People's Republic of China
| | - Chi Zhang
- Department of Cardiology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, Shandong, People's Republic of China
| | - Zhuoqi Li
- Radiotherapy Department, Shandong Second Provincial General Hospital, Shandong University, Jinan, 250299, Shandong, People's Republic of China
| | - Yuanyuan Wang
- Department of Oncology, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250299, Shandong, People's Republic of China
| | - Kai Zhang
- General Surgery Department, Wen-Shang County People's Hospital, Wenshang, 272500, Shandong, People's Republic of China
| | - Feng Chen
- Department of Thoracic Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, People's Republic of China
| | - Qi Dang
- Phase I Clinical Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University, and Shandong Academy of Medical Sciences, No. 440, Jiyan Road, Huaiyin District, Jinan City, 250117, Shandong, People's Republic of China.
| |
Collapse
|
205
|
Mok S, Ağaç Çobanoğlu D, Liu H, Mancuso JJ, Allison JP. Post-immunotherapy CTLA-4 Ig treatment improves antitumor efficacy. Proc Natl Acad Sci U S A 2024; 121:e2404661121. [PMID: 38923991 PMCID: PMC11228532 DOI: 10.1073/pnas.2404661121] [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: 03/05/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
Immune checkpoint therapies (ICT) improve overall survival of patients with cancer but may cause immune-related adverse events (irAEs) such as myocarditis. Cytotoxic T lymphocyte-associated antigen 4 immunoglobulin fusion protein (CTLA-4 Ig), an inhibitor of T cell costimulation through CD28, reverses irAEs in animal models. However, concerns exist about potentially compromising antitumor response of ICT. In mouse tumor models, we administered CTLA-4 Ig 1) concomitantly with ICT or 2) after ICT completion. Concomitant treatment reduced antitumor efficacy, while post-ICT administration improved efficacy without affecting frequency and function of CD8 T cells. The improved response was independent of the ICT used, whether CTLA-4 or PD-1 blockade. The frequency of Tregs was significantly decreased with CTLA-4 Ig. The resulting increased CD8/Treg ratio potentially underlies the enhanced efficacy of ICT followed by CTLA-4 Ig. This paradoxical mechanism shows that a CTLA-4 Ig regimen shown to reduce irAE severity does not compromise antitumor efficacy.
Collapse
Affiliation(s)
- Stephen Mok
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX77030
| | - Didem Ağaç Çobanoğlu
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX77030
| | - Huey Liu
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX77030
| | - James J. Mancuso
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX77030
- Parker Institute for Cancer Immunotherapy, The University of Texas MD Anderson Cancer Center, Houston, TX77030
| | - James P. Allison
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX77030
- Parker Institute for Cancer Immunotherapy, The University of Texas MD Anderson Cancer Center, Houston, TX77030
| |
Collapse
|
206
|
Chae YJ, Lee KG, Oh D, Lee SK, Park Y, Kim J. Antibody-Conjugated Nanogel with Two Immune Checkpoint Inhibitors for Enhanced Cancer Immunotherapy. Adv Healthc Mater 2024; 13:e2400235. [PMID: 38569198 DOI: 10.1002/adhm.202400235] [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: 01/20/2024] [Revised: 04/01/2024] [Indexed: 04/05/2024]
Abstract
Cancer immunotherapy by immune checkpoint inhibitors (ICIs) acts on antitumor responses by stimulating the immune system to attack cancer cells. However, this powerful therapy is hampered by its high treatment cost and limited efficacy. Here, it is shown that the development of an antibody-conjugated nanogel (ANGel), consisting of N-isopropylacrylamide-co-acrylic acid and antibody-binding protein (protein A), potentiates the efficacy of two ICI monoclonal antibodies (mAbs) (cytotoxic-T-lymphocyte-associated antigen 4 and programmed death ligand-1 mAbs). Compared with mAb treatment alone, treatment with a bispecific ANGel surface-conjugated with the mAbs significantly decreases both the survival of Michigan Cancer Foundation-7 (MCF-7) and M D Anderson-Metastatic Breast-231 (MDA-MB-231) breast cancer cells in vitro and the burden of 4T1-luciferase-2-derived orthotopic syngeneic tumors in vivo. The bispecific ANGel is also more potent than the conventional treatment at prolonging survival in animals with triple-negative breast cancer. The advantage of the bispecific ANGel over other engineered bispecific antibodies arises not only from the adaptability to link multiple antibodies quickly and easily, but also from the capability to maintain the anticancer effect steadily at subcutaneously delivered tumor site. This finding has an important implication for cancer immunotherapy, opening a new paradigm to treat solid tumors.
Collapse
Affiliation(s)
- Yun Jin Chae
- R&D Center, Scholar Foxtrot Co. Ltd., Seoul, 02796, Republic of Korea
| | - Kang-Gon Lee
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Doogie Oh
- R&D Center, Scholar Foxtrot Co. Ltd., Seoul, 02796, Republic of Korea
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Su-Kyoung Lee
- R&D Center, Scholar Foxtrot Co. Ltd., Seoul, 02796, Republic of Korea
| | - Yongdoo Park
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Jongseong Kim
- R&D Center, Scholar Foxtrot Co. Ltd., Seoul, 02796, Republic of Korea
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| |
Collapse
|
207
|
Zemek RM, Anagnostou V, Pires da Silva I, Long GV, Lesterhuis WJ. Exploiting temporal aspects of cancer immunotherapy. Nat Rev Cancer 2024; 24:480-497. [PMID: 38886574 DOI: 10.1038/s41568-024-00699-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/25/2024] [Indexed: 06/20/2024]
Abstract
Many mechanisms underlying an effective immunotherapy-induced antitumour response are transient and critically time dependent. This is equally true for several immunological events in the tumour microenvironment induced by other cancer treatments. Immune checkpoint therapy (ICT) has proven to be very effective in the treatment of some cancers, but unfortunately, with many cancer types, most patients do not experience a benefit. To improve outcomes, a multitude of clinical trials are testing combinations of ICT with various other treatment modalities. Ideally, those combination treatments should take time-dependent immunological events into account. Recent studies have started to map the dynamic cellular and molecular changes that occur during treatment with ICT, in the tumour and systemically. Here, we overlay the dynamic ICT response with the therapeutic response following surgery, radiotherapy, chemotherapy and targeted therapies. We propose that by combining treatments in a time-conscious manner, we may optimally exploit the interactions between the individual therapies.
Collapse
Affiliation(s)
- Rachael M Zemek
- Telethon Kids Institute, University of Western Australia, Perth, Western Australia, Australia
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Valsamo Anagnostou
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Inês Pires da Silva
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine & Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- Crown Princess Mary Cancer Centre Westmead, Blacktown Hospital, Sydney, New South Wales, Australia
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine & Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia
| | - Willem Joost Lesterhuis
- Telethon Kids Institute, University of Western Australia, Perth, Western Australia, Australia.
| |
Collapse
|
208
|
Holder AM, Dedeilia A, Sierra-Davidson K, Cohen S, Liu D, Parikh A, Boland GM. Defining clinically useful biomarkers of immune checkpoint inhibitors in solid tumours. Nat Rev Cancer 2024; 24:498-512. [PMID: 38867074 DOI: 10.1038/s41568-024-00705-7] [Citation(s) in RCA: 62] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/08/2024] [Indexed: 06/14/2024]
Abstract
Although more than a decade has passed since the approval of immune checkpoint inhibitors (ICIs) for the treatment of melanoma and non-small-cell lung, breast and gastrointestinal cancers, many patients still show limited response. US Food and Drug Administration (FDA)-approved biomarkers include programmed cell death 1 ligand 1 (PDL1) expression, microsatellite status (that is, microsatellite instability-high (MSI-H)) and tumour mutational burden (TMB), but these have limited utility and/or lack standardized testing approaches for pan-cancer applications. Tissue-based analytes (such as tumour gene signatures, tumour antigen presentation or tumour microenvironment profiles) show a correlation with immune response, but equally, these demonstrate limited efficacy, as they represent a single time point and a single spatial assessment. Patient heterogeneity as well as inter- and intra-tumoural differences across different tissue sites and time points represent substantial challenges for static biomarkers. However, dynamic biomarkers such as longitudinal biopsies or novel, less-invasive markers such as blood-based biomarkers, radiomics and the gut microbiome show increasing potential for the dynamic identification of ICI response, and patient-tailored predictors identified through neoadjuvant trials or novel ex vivo tumour models can help to personalize treatment. In this Perspective, we critically assess the multiple new static, dynamic and patient-specific biomarkers, highlight the newest consortia and trial efforts, and provide recommendations for future clinical trials to make meaningful steps forwards in the field.
Collapse
Affiliation(s)
- Ashley M Holder
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Sonia Cohen
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - David Liu
- Dana Farber Cancer Institute, Boston, MA, USA
| | - Aparna Parikh
- Cancer Center, Massachusetts General Hospital, Boston, MA, USA
| | - Genevieve M Boland
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA.
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
209
|
Zeng J, Fang Y, Zhang Z, Lv Z, Wang X, Huang Q, Tian Z, Li J, Xu W, Zhu W, Yu J, Liu T, Qian Q. Antitumor activity of Z15-0-2, a bispecific nanobody targeting PD-1 and CTLA-4. Oncogene 2024; 43:2244-2252. [PMID: 38806619 PMCID: PMC11245388 DOI: 10.1038/s41388-024-03066-5] [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: 03/06/2024] [Revised: 05/09/2024] [Accepted: 05/13/2024] [Indexed: 05/30/2024]
Abstract
The combination of programmed cell death protein 1 (PD-1) and cytotoxic T lymphocyte antigen 4 (CTLA-4) antibodies has potential for enhancing clinical efficacy. We described the development and antitumor activity of Z15-0, a bispecific nanobody targeting both the PD-1 and CTLA-4 pathways simultaneously. We designed and optimized the mRNA sequence encoding Z15-0, referred to as Z15-0-2 and through a series of in vitro and in vivo experiments, we established that the optimized Z15-0-2 mRNA sequence significantly increased the expression of the bispecific nanobody. Administration of Z15-0-2 mRNA to tumor-bearing mice led to greater inhibition of tumor growth compared to controls. In aggregate, we introduced a novel bispecific nanobody and have re-engineered it to boost expression of mRNA, representing a new drug development paradigm.
Collapse
Affiliation(s)
- Jianyao Zeng
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Yuan Fang
- Shanghai Cell Therapy Group Co., Ltd, Shanghai, 201805, China
| | - Zixuan Zhang
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Zhenzhen Lv
- Shanghai Cell Therapy Group Co., Ltd, Shanghai, 201805, China
| | - Xiaodie Wang
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Qian Huang
- Shanghai Cell Therapy Group Co., Ltd, Shanghai, 201805, China
| | - Zhidan Tian
- Shanghai Cell Therapy Group Co., Ltd, Shanghai, 201805, China
| | - Jiaguo Li
- Shanghai Cell Therapy Group Co., Ltd, Shanghai, 201805, China
| | - Wenfeng Xu
- Shanghai Cell Therapy Group Co., Ltd, Shanghai, 201805, China
| | - Weimin Zhu
- Shanghai Cell Therapy Group Co., Ltd, Shanghai, 201805, China
| | - Jing Yu
- Shanghai Cell Therapy Group Co., Ltd, Shanghai, 201805, China
| | - Tao Liu
- Shanghai Cell Therapy Group Co., Ltd, Shanghai, 201805, China.
| | - Qijun Qian
- School of Medicine, Shanghai University, Shanghai, 200444, China.
- Shanghai Cell Therapy Group Co., Ltd, Shanghai, 201805, China.
- Shanghai Mengchao Cancer Hospital, Shanghai University, Shanghai, 201805, China.
| |
Collapse
|
210
|
Gergely TG, Drobni ZD, Kallikourdis M, Zhu H, Meijers WC, Neilan TG, Rassaf T, Ferdinandy P, Varga ZV. Immune checkpoints in cardiac physiology and pathology: therapeutic targets for heart failure. Nat Rev Cardiol 2024; 21:443-462. [PMID: 38279046 DOI: 10.1038/s41569-023-00986-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/12/2023] [Indexed: 01/28/2024]
Abstract
Immune checkpoint molecules are physiological regulators of the adaptive immune response. Immune checkpoint inhibitors (ICIs), such as monoclonal antibodies targeting programmed cell death protein 1 or cytotoxic T lymphocyte-associated protein 4, have revolutionized cancer treatment and their clinical use is increasing. However, ICIs can cause various immune-related adverse events, including acute and chronic cardiotoxicity. Of these cardiovascular complications, ICI-induced acute fulminant myocarditis is the most studied, although emerging clinical and preclinical data are uncovering the importance of other ICI-related chronic cardiovascular complications, such as accelerated atherosclerosis and non-myocarditis-related heart failure. These complications could be more difficult to diagnose, given that they might only be present alongside other comorbidities. The occurrence of these complications suggests a potential role of immune checkpoint molecules in maintaining cardiovascular homeostasis, and disruption of physiological immune checkpoint signalling might thus lead to cardiac pathologies, including heart failure. Although inflammation is a long-known contributor to the development of heart failure, the therapeutic targeting of pro-inflammatory pathways has not been successful thus far. The increasingly recognized role of immune checkpoint molecules in the failing heart highlights their potential use as immunotherapeutic targets for heart failure. In this Review, we summarize the available data on ICI-induced cardiac dysfunction and heart failure, and discuss how immune checkpoint signalling is altered in the failing heart. Furthermore, we describe how pharmacological targeting of immune checkpoints could be used to treat heart failure.
Collapse
Affiliation(s)
- Tamás G Gergely
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | - Zsófia D Drobni
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Marinos Kallikourdis
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Adaptive Immunity Lab, Humanitas Research Hospital IRCCS, Milan, Italy
| | - Han Zhu
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Wouter C Meijers
- Erasmus MC, Cardiovascular Institute, Thorax Center, Department of Cardiology, Rotterdam, The Netherlands
| | - Tomas G Neilan
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Tienush Rassaf
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center Essen, Medical Faculty, University Hospital Essen, Essen, Germany
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Zoltán V Varga
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary.
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary.
| |
Collapse
|
211
|
Liu Q, Ma F, Zhang G. Molecular subtyping of skin cutaneous melanoma based on inflammatory response. Heliyon 2024; 10:e33088. [PMID: 39005905 PMCID: PMC11239590 DOI: 10.1016/j.heliyon.2024.e33088] [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: 12/21/2023] [Revised: 06/03/2024] [Accepted: 06/13/2024] [Indexed: 07/16/2024] Open
Abstract
The inflammatory response plays a crucial role in determining the prognosis and therapeutic response of skin cutaneous melanoma (SKCM). However, the molecular subtypes based on the inflammatory response and their clinical significance in SKCM have not been extensively studied. Clustering analyses to identify inflammation subtypes of SKCM based on the expression levels of inflammation response gene. We identified three subtypes: Inflammation_H, Inflammation_M, and Inflammation_L, which offer a more nuanced understanding of the complex relationship between inflammation and SKCM. The Inflammation_H subtype is associated with the most favourable prognosis, and is characterised by high levels of immune infiltrates and PD-L1 expression, low levels of stemness, high differentiation, and high genomic stability. In contrast, the Inflammation_L subtype has the least favourable prognosis, with the lowest levels of immune infiltrates and PD-L1 expression, high levels of stemness, low differentiation, and low genomic stability. In addition, the Inf-score, which is a linear risk scoring model based on the expression levels of inflammatory response genes, can be a useful tool for clinicians to assess SKCM prognosis and guide therapeutic decisions. This scoring model shows promise for clinical use in predicting patient outcomes and helps clinicians tailor treatments for individual patients. In conclusion, these findings represent a significant contribution to our understanding of the molecular subtypes of SKCM based on the levels of inflammatory response genes and their potential clinical significance. However, further studies are necessary to validate these findings and explore the underlying mechanisms of different subtypes.
Collapse
Affiliation(s)
- Qian Liu
- Big Data Institute, Central South University, Changsha, Hunan, China
| | - Fangyu Ma
- Health Management center Xiangya Hospital, Central South University, China
| | - Guanxiong Zhang
- The Department of Dermatology, Xiangya Hospital, Central South University, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China
- Furong Laboratory, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, China
| |
Collapse
|
212
|
Shannon AB, Zager JS, Perez MC. Clinical Characteristics and Special Considerations in the Management of Rare Melanoma Subtypes. Cancers (Basel) 2024; 16:2395. [PMID: 39001457 PMCID: PMC11240680 DOI: 10.3390/cancers16132395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
Rare histologic subtypes of melanoma, including acral, mucosal, uveal, and desmoplastic melanomas, only make up 5% of all diagnosed melanomas and are often underrepresented in large, randomized trials. Recent advancements in systemic therapy have shown marked improvement in pathologic response rates, improving progression-free and overall survival among cutaneous melanoma patients, but there are limited data to demonstrate improved survival among rarer subtypes of melanoma. Acral melanoma has a poor response to immunotherapy and is associated with worse survival. Mucosal melanoma has a large variability in its presentation, a poor prognosis, and a low mutational burden. Uveal melanoma is associated with a high rate of liver metastasis; recent adoption of infusion and perfusion therapies has demonstrated improved survival among these patients. Desmoplastic melanoma, a high-risk cutaneous melanoma, is associated with high locoregional recurrence rates and mutational burden, suggesting this melanoma may have enhanced response to immunotherapy. While these variants of melanoma represent distinct disease entities, this review highlights the clinicopathologic characteristics and treatment recommendations for each of these rare melanomas and highlights the utility of modern therapies for each of them.
Collapse
Affiliation(s)
- Adrienne B Shannon
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Jonathan S Zager
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Matthew C Perez
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| |
Collapse
|
213
|
Guzik P, Łukasiewicz M, Harpula M, Zając P, Żmuda M, Śniadecki M, Topolewski P. Survival and Treatment Modalities in Primary Vaginal Melanoma-Case Report and a Narrative Review. J Clin Med 2024; 13:3771. [PMID: 38999339 PMCID: PMC11242499 DOI: 10.3390/jcm13133771] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/03/2024] [Accepted: 06/13/2024] [Indexed: 07/14/2024] Open
Abstract
Background/Objectives: Primary vaginal melanoma (PVM) is a rare cancer representing five percent of vaginal cancers and less than one percent of all female vaginal melanomas, with an incidence rate of 0.46 per million women per year. The aim of this study was to present a case of combined therapy and conservative surgical treatment in a young patient with PVM and to perform a systematic review of the same subject. Methods: We performed a narrative review of the literature and presented a case report. Results: The review yielded a total of 43 articles. We presented treatment modalities and survival outcomes. The presented case involved a combination of surgical treatment with adjuvant therapy comprising nivolumab and ipilimumab. Conclusions: PVM is a disease with a poor prognosis; however, new treatment options are promising and have a great chance of significantly improving survival. The combination of the wide local excision of the primary lesion followed by adjuvant therapies results in the best outcomes in the treatment of PVM. Future clinical studies are warranted to provide new evidence for the treatment outcomes of nonsurgical, metastatic PVM and the adjuvant treatment of PVM.
Collapse
Affiliation(s)
- Paweł Guzik
- Clinical Department of Gynecology and Obstetrics, City Hospital, 35-241 Rzeszów, Poland; (M.H.); (P.Z.)
| | - Martyna Łukasiewicz
- Medical University of Gdańsk, 17 Smoluchowskiego St., 80-241 Gdańsk, Poland; (M.Ł.); (P.T.)
| | - Magdalena Harpula
- Clinical Department of Gynecology and Obstetrics, City Hospital, 35-241 Rzeszów, Poland; (M.H.); (P.Z.)
| | - Paweł Zając
- Clinical Department of Gynecology and Obstetrics, City Hospital, 35-241 Rzeszów, Poland; (M.H.); (P.Z.)
| | - Marcin Żmuda
- Pathology Department, Clinical Provincial Hospital no 2, 35-241 Rzeszów, Poland;
| | - Marcin Śniadecki
- Department of Gynecology and Obstetrics, Medical University of Gdańsk, 17 Smoluchowskiego St., 80-241 Gdańsk, Poland;
| | - Paweł Topolewski
- Medical University of Gdańsk, 17 Smoluchowskiego St., 80-241 Gdańsk, Poland; (M.Ł.); (P.T.)
| |
Collapse
|
214
|
Poalelungi DG, Neagu AI, Fulga A, Neagu M, Tutunaru D, Nechita A, Fulga I. Revolutionizing Pathology with Artificial Intelligence: Innovations in Immunohistochemistry. J Pers Med 2024; 14:693. [PMID: 39063947 PMCID: PMC11278211 DOI: 10.3390/jpm14070693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
Artificial intelligence (AI) is a reality of our times, and it has been successfully implemented in all fields, including medicine. As a relatively new domain, all efforts are directed towards creating algorithms applicable in most medical specialties. Pathology, as one of the most important areas of interest for precision medicine, has received significant attention in the development and implementation of AI algorithms. This focus is especially important for achieving accurate diagnoses. Moreover, immunohistochemistry (IHC) serves as a complementary diagnostic tool in pathology. It can be further augmented through the application of deep learning (DL) and machine learning (ML) algorithms for assessing and analyzing immunohistochemical markers. Such advancements can aid in delineating targeted therapeutic approaches and prognostic stratification. This article explores the applications and integration of various AI software programs and platforms used in immunohistochemical analysis. It concludes by highlighting the application of these technologies to pathologies such as breast, prostate, lung, melanocytic proliferations, and hematologic conditions. Additionally, it underscores the necessity for further innovative diagnostic algorithms to assist physicians in the diagnostic process.
Collapse
Affiliation(s)
- Diana Gina Poalelungi
- Faculty of Medicine and Pharmacy, Dunarea de Jos University of Galati, 35 AI Cuza St., 800010 Galati, Romania; (D.G.P.); (M.N.); (D.T.); (A.N.); (I.F.)
- Saint Apostle Andrew Emergency County Clinical Hospital, 177 Brailei St., 800578 Galati, Romania
| | - Anca Iulia Neagu
- Faculty of Medicine and Pharmacy, Dunarea de Jos University of Galati, 35 AI Cuza St., 800010 Galati, Romania; (D.G.P.); (M.N.); (D.T.); (A.N.); (I.F.)
- Saint John Clinical Emergency Hospital for Children, 800487 Galati, Romania
| | - Ana Fulga
- Faculty of Medicine and Pharmacy, Dunarea de Jos University of Galati, 35 AI Cuza St., 800010 Galati, Romania; (D.G.P.); (M.N.); (D.T.); (A.N.); (I.F.)
- Saint Apostle Andrew Emergency County Clinical Hospital, 177 Brailei St., 800578 Galati, Romania
| | - Marius Neagu
- Faculty of Medicine and Pharmacy, Dunarea de Jos University of Galati, 35 AI Cuza St., 800010 Galati, Romania; (D.G.P.); (M.N.); (D.T.); (A.N.); (I.F.)
- Saint Apostle Andrew Emergency County Clinical Hospital, 177 Brailei St., 800578 Galati, Romania
| | - Dana Tutunaru
- Faculty of Medicine and Pharmacy, Dunarea de Jos University of Galati, 35 AI Cuza St., 800010 Galati, Romania; (D.G.P.); (M.N.); (D.T.); (A.N.); (I.F.)
- Saint Apostle Andrew Emergency County Clinical Hospital, 177 Brailei St., 800578 Galati, Romania
| | - Aurel Nechita
- Faculty of Medicine and Pharmacy, Dunarea de Jos University of Galati, 35 AI Cuza St., 800010 Galati, Romania; (D.G.P.); (M.N.); (D.T.); (A.N.); (I.F.)
- Saint John Clinical Emergency Hospital for Children, 800487 Galati, Romania
| | - Iuliu Fulga
- Faculty of Medicine and Pharmacy, Dunarea de Jos University of Galati, 35 AI Cuza St., 800010 Galati, Romania; (D.G.P.); (M.N.); (D.T.); (A.N.); (I.F.)
- Saint Apostle Andrew Emergency County Clinical Hospital, 177 Brailei St., 800578 Galati, Romania
| |
Collapse
|
215
|
Quandt Z, Jacob S, Fadlullah MZH, Wu C, Wu C, Huppert L, Levine LS, Sison P, Tsai KK, Chow M, Kang JH, Hwang J, Lee JC, Oglesby A, Venegas J, Brintz BJ, Tan AC, Anderson MS, Rosenblum MD, Young A, Daud AI. Phase II trial of pembrolizumab, ipilimumab, and aspirin in melanoma: clinical outcomes and translational predictors of response. BJC REPORTS 2024; 2:46. [PMID: 39516257 PMCID: PMC11524064 DOI: 10.1038/s44276-024-00057-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/26/2024] [Accepted: 03/08/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVE Many patients with melanoma treated with immune checkpoint inhibitors (ICIs) do not derive response. Preclinical and retrospective studies identified that inhibition of the cyclooxygenase (COX) pathway may improve response to ICI treatment. METHODS This prospective single site phase II trial accrued patients with advanced/metastatic melanoma. Participants underwent high-dose aspirin daily combined with pembrolizumab and ipilimumab every 3 weeks for 4 cycles followed by high-dose aspirin and pembrolizumab monotherapy. The primary endpoint was objective response rate (ORR). Longitudinal sampling of blood was performed to assess peripheral immune correlates. RESULTS Twenty-seven subjects were enrolled with median follow-up of 32 months. An ORR of 62.9% was reached prior to discontinuation due to low likelihood of achieving the pre-specified ORR of 80%. 17 patients (63%) experienced a treatment-related adverse event (TRAEs) grade 3 or higher. A per-protocol analysis showed that patients able to continue aspirin alongside ICI through the induction period experienced significant survival benefit. Ten cytokines and increased regulatory T cells in the periphery correlated with beneficial response. CONCLUSIONS The addition of high-dose aspirin to combination ICI within this study results in response comparable to ICI alone. Future clinical studies of COX inhibition will need to focus on mitigation of AEs to establish the clinical utility of this combination.
Collapse
Affiliation(s)
- Zoe Quandt
- Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Diabetes Center, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Saya Jacob
- Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, 94143, USA
| | | | - Chaorong Wu
- Division of Epidemiology, University of Utah, Salt Lake City, UT, 84112, USA
| | - Clinton Wu
- Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Laura Huppert
- Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Lauren S Levine
- Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Paula Sison
- Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Katy K Tsai
- Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Melissa Chow
- Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Jee Hye Kang
- Diabetes Center, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Jimmy Hwang
- Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, 94143, USA
| | - James C Lee
- Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Ariel Oglesby
- Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Jessica Venegas
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, UT, 84112, USA
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, 84112, USA
| | - Ben J Brintz
- Division of Epidemiology, University of Utah, Salt Lake City, UT, 84112, USA
| | - Aik Choon Tan
- Departments of Oncological Sciences and Biomedical Informatics, University of Utah, Salt Lake City, UT, 84112, USA
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, UT, 84112, USA
| | - Mark S Anderson
- Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Diabetes Center, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Michael D Rosenblum
- Dermatology, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Arabella Young
- Diabetes Center, University of California San Francisco, San Francisco, CA, 94143, USA.
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, UT, 84112, USA.
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, 84112, USA.
| | - Adil I Daud
- Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA.
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, 94143, USA.
| |
Collapse
|
216
|
Lorenzo-Sanz L, Lopez-Cerda M, da Silva-Diz V, Artés MH, Llop S, Penin RM, Bermejo JO, Gonzalez-Suarez E, Esteller M, Viñals F, Espinosa E, Oliva M, Piulats JM, Martin-Liberal J, Muñoz P. Cancer cell plasticity defines response to immunotherapy in cutaneous squamous cell carcinoma. Nat Commun 2024; 15:5352. [PMID: 38914547 PMCID: PMC11196727 DOI: 10.1038/s41467-024-49718-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 06/17/2024] [Indexed: 06/26/2024] Open
Abstract
Immune checkpoint blockade (ICB) approaches have changed the therapeutic landscape for many tumor types. However, half of cutaneous squamous cell carcinoma (cSCC) patients remain unresponsive or develop resistance. Here, we show that, during cSCC progression in male mice, cancer cells acquire epithelial/mesenchymal plasticity and change their immune checkpoint (IC) ligand profile according to their features, dictating the IC pathways involved in immune evasion. Epithelial cancer cells, through the PD-1/PD-L1 pathway, and mesenchymal cancer cells, through the CTLA-4/CD80 and TIGIT/CD155 pathways, differentially block antitumor immune responses and determine the response to ICB therapies. Accordingly, the anti-PD-L1/TIGIT combination is the most effective strategy for blocking the growth of cSCCs that contain both epithelial and mesenchymal cancer cells. The expression of E-cadherin/Vimentin/CD80/CD155 proteins in cSCC, HNSCC and melanoma patient samples predicts response to anti-PD-1/PD-L1 therapy. Collectively, our findings indicate that the selection of ICB therapies should take into account the epithelial/mesenchymal features of cancer cells.
Collapse
Affiliation(s)
- Laura Lorenzo-Sanz
- Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), 08908, L'Hospitalet de Llobregat, Barcelona, Spain.
| | - Marta Lopez-Cerda
- Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), 08908, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Victoria da Silva-Diz
- Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), 08908, L'Hospitalet de Llobregat, Barcelona, Spain
- Rutgers Cancer Institute of New Jersey, Rutgers University, 08901, New Brunswick, NJ, USA
| | - Marta H Artés
- Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), 08908, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Sandra Llop
- Medical Oncology Department, Catalan Institute of Oncology (ICO), 08908, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Rosa M Penin
- Pathology Service, Bellvitge University Hospital/IDIBELL, 08908, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Josep Oriol Bermejo
- Plastic Surgery Unit, Bellvitge University Hospital/IDIBELL, 08908, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Eva Gonzalez-Suarez
- Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), 08908, L'Hospitalet de Llobregat, Barcelona, Spain
- Molecular Oncology, Spanish National Cancer Research Centre (CNIO), 28029, Madrid, Spain
| | - Manel Esteller
- Josep Carreras Leukaemia Research Institute (IJC), 08916, Badalona, Barcelona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), ISCIII, 28029, Madrid, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010, Barcelona, Spain
- Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona (UB), 08908, Barcelona, Spain
| | - Francesc Viñals
- Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), 08908, L'Hospitalet de Llobregat, Barcelona, Spain
- Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona (UB), 08908, Barcelona, Spain
- Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology (ICO)/IDIBELL, 08908, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Enrique Espinosa
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), ISCIII, 28029, Madrid, Spain
- Medical Oncology Department, La Paz University Hospital, Autonomous University of Madrid (UAM), 28046, Madrid, Spain
| | - Marc Oliva
- Medical Oncology Department, Catalan Institute of Oncology (ICO), 08908, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Josep M Piulats
- Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), 08908, L'Hospitalet de Llobregat, Barcelona, Spain
- Medical Oncology Department, Catalan Institute of Oncology (ICO), 08908, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Juan Martin-Liberal
- Medical Oncology Department, Catalan Institute of Oncology (ICO), 08908, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Purificación Muñoz
- Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), 08908, L'Hospitalet de Llobregat, Barcelona, Spain.
| |
Collapse
|
217
|
Xu H, Zhao X, Luo J. Combination of tumor antigen drainage and immune activation to promote a cancer-immunity cycle against glioblastoma. Cell Mol Life Sci 2024; 81:275. [PMID: 38907858 PMCID: PMC11335198 DOI: 10.1007/s00018-024-05300-5] [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: 01/02/2024] [Revised: 04/26/2024] [Accepted: 05/28/2024] [Indexed: 06/24/2024]
Abstract
While conventional cancer modalities, such as chemotherapy and radiotherapy, act through direct killing of tumor cells, cancer immunotherapy elicits potent anti-tumor immune responses thereby eliminating tumors. Nevertheless, promising outcomes have not been reported in patients with glioblastoma (GBM) likely due to the immune privileged status of the central nervous system and immunosuppressive micro-environment within GBM. In the past years, several exciting findings, such as the re-discovery of meningeal lymphatic vessels (MLVs), three-dimensional anatomical reconstruction of MLV networks, and the demonstration of the promotion of GBM immunosurveillance by lymphatic drainage enhancement, have revealed an intricate communication between the nervous and immune systems, and brought hope for the development of new GBM treatment. Based on conceptual framework of the updated cancer-immunity (CI) cycle, here we focus on GBM antigen drainage and immune activation, the early events in driving the CI cycle. We also discuss the implications of these findings for developing new therapeutic approaches in tackling fatal GBM in the future.
Collapse
Affiliation(s)
- Han Xu
- Laboratory of Vascular Biology, Institute of Molecular Medicine, College of Future Technology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, 100871, China
| | - Xiaomei Zhao
- Laboratory of Vascular Biology, Institute of Molecular Medicine, College of Future Technology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, 100871, China
| | - Jincai Luo
- Laboratory of Vascular Biology, Institute of Molecular Medicine, College of Future Technology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, 100871, China.
| |
Collapse
|
218
|
Shah NJ, Della Pia A, Wu T, Williams A, Weber M, Sinclaire B, Gourna Paleoudis E, Alaoui A, Lev-Ari S, Adams S, Kaufman J, Parikh SB, Tonti E, Muller E, Serzan M, Cheruku D, Lee A, Sridhar A, Hee B(TP, Ahn J, Pecora A, Ip A, Atkins MB. Clinical Outcomes of Immune Checkpoint Inhibitors in Unique Cohorts Underrepresented in Clinical Trials. Cancers (Basel) 2024; 16:2223. [PMID: 38927928 PMCID: PMC11202168 DOI: 10.3390/cancers16122223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/29/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Regulatory approval of immune checkpoint inhibitors (ICIs) was based on results of large, randomized clinical trials, resulting in limited outcomes data in patient cohorts typically underrepresented in such trials. The objective of this study was to evaluate the efficacy and safety of ICIs in these unique patient cohorts. This is a multicenter, retrospective analysis of real-world data at six academic and community clinics in the United States from 1 January 2011 to 1 April 2018. Patients were included if they had received at least one cycle of ICI treatment. Unique patient cohorts included age > 75 years, non-White race, positive smoking history, ECOG performance status (PS) ≥ 2, BMI ≥ 30 kg/m2, autoimmune diseases (AIDs), chronic viral infections (CVI), extensive prior lines of therapy (LOTs), or >three metastatic sites. Immune-related adverse events (irAEs), overall survival (OS), and time to treatment failure were evaluated in the entire cohort and in NSCLC patients treated with PD-(L)1 monotherapy. Outcomes and their association with unique patient cohorts were compared on univariate analysis and multivariate analysis to those without a particular characteristic in the entire NSCLC PD-(L)1 monotherapy cohorts. In total, 1453 patients were included: 56.5%-smokers, 30.4%-non-White, 22.8%-elderly, 20.8%-ECOG PS ≥ 2, 15.7%-history of AIDs, and 4.7%-history of CVI. The common ICIs were nivolumab (37.1%) and pembrolizumab (22.2%). Black patients, compared to White patients, experienced fewer irAEs (OR 0.54, p < 0.001). An ECOG PS of ≥2 (HR = 2.01, p < 0.001) and an increased number of previous LOTs were associated with poor OS (the median OS of 26.2 vs. 16.2 vs. 9.6 months for one vs. two vs. three prior LOTs, p < 0.001). The above results were confirmed in anti-PD-(L)1 monotherapy non-small cell lung cancer patients (n = 384). Overall, ICIs were safe and efficacious in these typically underrepresented patient cohorts. We noted ECOG PS ≥ 2 and an increased prior LOTs were associated with poor ICI efficacy, and Black patients, compared to White patients, experienced fewer irAEs.
Collapse
Affiliation(s)
- Neil J. Shah
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Department of Medicine, Weill Cornell Medical Center, New York, NY 10065, USA
| | - Alexandra Della Pia
- John Theurer Cancer Center at Hackensack Meridian Health, Hackensack, NJ 07601, USA
| | - Tianmin Wu
- Department of Biostatistics, Georgetown University Medical Center, Washington, DC 20007, USA
| | - Aquino Williams
- Hackensack Meridian Health Mountainside Medical Center, Montclair, NJ 07042, USA
| | - Melinda Weber
- John Theurer Cancer Center at Hackensack Meridian Health, Hackensack, NJ 07601, USA
| | - Brittany Sinclaire
- John Theurer Cancer Center at Hackensack Meridian Health, Hackensack, NJ 07601, USA
| | | | - Adil Alaoui
- Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Washington, DC 20007, USA
| | - Shaked Lev-Ari
- Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Washington, DC 20007, USA
| | - Shari Adams
- John Theurer Cancer Center at Hackensack Meridian Health, Hackensack, NJ 07601, USA
| | - Jordan Kaufman
- John Theurer Cancer Center at Hackensack Meridian Health, Hackensack, NJ 07601, USA
| | - Sahil B. Parikh
- John Theurer Cancer Center at Hackensack Meridian Health, Hackensack, NJ 07601, USA
| | - Emily Tonti
- John Theurer Cancer Center at Hackensack Meridian Health, Hackensack, NJ 07601, USA
| | - Eric Muller
- John Theurer Cancer Center at Hackensack Meridian Health, Hackensack, NJ 07601, USA
| | - Michael Serzan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Divya Cheruku
- Hackensack Meridian Health Mountainside Medical Center, Montclair, NJ 07042, USA
| | - Albert Lee
- Hackensack Meridian School of Medicine, Nutley, NJ 07110, USA
| | | | | | - Jaeil Ahn
- Department of Biostatistics, Georgetown University Medical Center, Washington, DC 20007, USA
| | - Andrew Pecora
- John Theurer Cancer Center at Hackensack Meridian Health, Hackensack, NJ 07601, USA
- Hackensack Meridian School of Medicine, Nutley, NJ 07110, USA
| | - Andrew Ip
- John Theurer Cancer Center at Hackensack Meridian Health, Hackensack, NJ 07601, USA
- Hackensack Meridian School of Medicine, Nutley, NJ 07110, USA
| | - Michael B. Atkins
- Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Washington, DC 20007, USA
| |
Collapse
|
219
|
Martinez-Cannon BA, Colombo I. The evolving role of immune checkpoint inhibitors in cervical and endometrial cancer. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:23. [PMID: 39050882 PMCID: PMC11267150 DOI: 10.20517/cdr.2023.120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 04/24/2024] [Accepted: 06/04/2024] [Indexed: 07/27/2024]
Abstract
The introduction of immune checkpoint inhibitors (ICIs) has revolutionized the treatment landscape for numerous tumor types, including cervical and endometrial cancers. Multiple ICIs against programmed cell death-1 (PD-1), programmed death-ligand 1 (PD-L1), and cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) have demonstrated encouraging outcomes in controlled clinical studies for advanced cervical and endometrial cancers. For advanced cervical cancer, approved ICIs as second-line treatment include cemiplimab, nivolumab, and pembrolizumab as single agents. In the first-line treatment setting, options include pembrolizumab alone or in combination with bevacizumab, as well as atezolizumab combined with a backbone platinum-based chemotherapy plus bevacizumab. Additionally, for locally advanced cervical cancer, pembrolizumab is recommended alongside concurrent chemoradiotherapy. For endometrial cancer, pembrolizumab monotherapy, pembrolizumab in combination with lenvatinib, and dostarlimab are currently approved as second-line treatment options. Moreover, either dostarlimab or pembrolizumab can be added to first-line platinum-based chemotherapy for mismatch repair deficient malignancies. Although the inclusion of these agents in clinical practice has led to improved overall response rates and survival outcomes, many patients still lack benefits, possibly due to multiple intrinsic and adaptive resistance mechanisms to immunotherapy. This review aims to highlight the rationale for utilizing ICIs and their current role, while also delineating the proposed mechanisms of resistance to ICIs in cervical and endometrial cancer.
Collapse
Affiliation(s)
- Bertha Alejandra Martinez-Cannon
- Hematology-Oncology Department, National Institute of Medical Sciences and Nutrition Salvador Zubiran, Mexico City 14080, Mexico
| | - Ilaria Colombo
- Medical Oncology, Oncology Institute of Southern Switzerland (IOSI), Ente Ospedaliero Cantonale (EOC), Bellinzona 6500 - CH, Switzerland
| |
Collapse
|
220
|
Hegewisch-Becker S, Mendez G, Chao J, Nemecek R, Feeney K, Van Cutsem E, Al-Batran SE, Mansoor W, Maisey N, Pazo Cid R, Burge M, Perez-Callejo D, Hipkin RW, Mukherjee S, Lei M, Tang H, Suryawanshi S, Kelly RJ, Tebbutt NC. First-Line Nivolumab and Relatlimab Plus Chemotherapy for Gastric or Gastroesophageal Junction Adenocarcinoma: The Phase II RELATIVITY-060 Study. J Clin Oncol 2024; 42:2080-2093. [PMID: 38723227 PMCID: PMC11191068 DOI: 10.1200/jco.23.01636] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 01/29/2024] [Accepted: 03/05/2024] [Indexed: 06/07/2024] Open
Abstract
PURPOSE Open-label phase II study (RELATIVITY-060) to investigate the efficacy and safety of first-line nivolumab, a PD-1-blocking antibody, plus relatlimab, a lymphocyte-activation gene 3 (LAG-3)-blocking antibody, plus chemotherapy in patients with previously untreated advanced gastric cancer (GC) or gastroesophageal junction cancer (GEJC). METHODS Patients with unresectable, locally advanced or metastatic GC/GEJC were randomly assigned 1:1 to nivolumab + relatlimab (fixed-dose combination) + chemotherapy or nivolumab + chemotherapy. The primary end point was objective response rate (ORR; per RECIST v1.1 by blinded independent central review [BICR]) in patients whose tumors had LAG-3 expression ≥1%. RESULTS Of 274 patients, 138 were randomly assigned to nivolumab + relatlimab + chemotherapy and 136 to nivolumab + chemotherapy. Median follow-up was 11.9 months. In patients with LAG-3 expression ≥1%, BICR-assessed ORR (95% CI) was 48% (38 to 59) in the nivolumab + relatlimab + chemotherapy arm and 61% (51 to 71) in the nivolumab + chemotherapy arm; median progression-free survival (95% CI) by BICR was 7.0 months (5.8 to 8.4) versus 8.3 months (6.9 to 12.1; hazard ratio [HR], 1.41 [95% CI, 0.97 to 2.05]), and median overall survival (95% CI) was 13.5 months (11.9 to 19.1) versus 16.0 months (10.9 to not estimable; HR, 1.04 [95% CI, 0.70 to 1.54]), respectively. Grade 3 or 4 treatment-related adverse events (TRAEs) occurred in 69% and 61% of all treated patients, and 42% and 36% of patients discontinued because of any-grade TRAEs in the nivolumab + relatlimab + chemotherapy and nivolumab + chemotherapy arms, respectively. CONCLUSION RELATIVITY-060 did not meet its primary end point of improved ORR in patients with LAG-3 expression ≥1% when relatlimab was added to nivolumab + chemotherapy compared with nivolumab + chemotherapy. Further studies are needed to address whether adding anti-LAG-3 to anti-PD-1 plus chemotherapy can benefit specific GC/GEJC patient subgroups.
Collapse
Affiliation(s)
| | - Guillermo Mendez
- Hospital Universitario Fundacion Favaloro, Buenos Aires, Argentina
| | - Joseph Chao
- City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Radim Nemecek
- Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Kynan Feeney
- St John of God Murdoch Hospital, Murdoch, WA, Australia
| | - Eric Van Cutsem
- University Hospitals Gasthuisberg and University of Leuven (KUL), Leuven, Belgium
| | - Salah-Eddin Al-Batran
- Krankenhaus Nordwest University Cancer Center Frankfurt, and Institut für Klinische Krebsforschung IKF am Krankenhaus Nordwest, Frankfurt, Germany
| | - Wasat Mansoor
- The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Nicholas Maisey
- Guy's and St Thomas's NHS Foundation Trust, London, United Kingdom
| | | | - Matthew Burge
- Royal Brisbane & Womens Hospital, Herston, QLD, Australia
| | | | | | | | - Ming Lei
- Bristol Myers Squibb, Princeton, NJ
| | - Hao Tang
- Bristol Myers Squibb, Princeton, NJ
| | | | | | - Niall C. Tebbutt
- Austin Health, Heidelberg, VIC, Australia
- University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
221
|
Mallardo D, Sparano F, Vitale MG, Trojaniello C, Fordellone M, Cioli E, Esposito A, Festino L, Mallardo M, Vanella V, Facchini BA, De Filippi R, Meinardi P, Ottaviano M, Caracò C, Simeone E, Ascierto PA. Impact of cemiplimab treatment duration on clinical outcomes in advanced cutaneous squamous cell carcinoma. Cancer Immunol Immunother 2024; 73:160. [PMID: 38850335 PMCID: PMC11162402 DOI: 10.1007/s00262-024-03728-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 05/10/2024] [Indexed: 06/10/2024]
Abstract
Treatment duration with checkpoint inhibitors must be optimized to prevent unjustified toxicity, but evidence for the management of cutaneous squamous cell carcinoma is lacking. A retrospective study was performed to evaluate the survival of patients with cutaneous squamous cell carcinoma (CSCC) who discontinued cemiplimab due to different causes and without progression. Among 95 patients with CSCC who received cemiplimab, 22 (23%) patients discontinued immunotherapy due to causes other than progression, such as comorbidities, toxicity, complete response or lack of compliance (group that discontinued before censoring [DBC]), then 73 patients had standard treatment scheduled (STS). The overall survival was 25.2 months (95% CI: 8.9-29.4) in STS group and 28.3 months (95% CI: 12.7-28.3) in the DBC group; deaths for all causes were 11/22 (50%) in the DBC group and 34/73 (46.6%) in the STS group (p = 0.32). 10/22 (45.4%) subjects died due to CSCC in the DBC after discontinuation and 34/73 (46.6%) in the STS group, and the difference between groups was not significant (p = 0.230). Duration of treatment was significantly lower in subjects with stable disease versus those with complete or partial response (16.9, 30.6 and 34.9 months, respectively; p = 0.004). Among the 22 STS patients, 12 received cemiplimab for less than 12 months (10 [83%] died) and 10 for at least 12 months (1 [10%] died). Our observation, finding no outcome difference between DBC and STS groups, suggests that ICI treatment after one year might expose patients to further treatment related events without efficacy advantages.
Collapse
MESH Headings
- Humans
- Male
- Female
- Skin Neoplasms/drug therapy
- Skin Neoplasms/mortality
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/mortality
- Aged
- Retrospective Studies
- Middle Aged
- Aged, 80 and over
- Treatment Outcome
- Antineoplastic Agents, Immunological/therapeutic use
- Antineoplastic Agents, Immunological/administration & dosage
- Antineoplastic Agents, Immunological/adverse effects
- Adult
- Immune Checkpoint Inhibitors/therapeutic use
- Immune Checkpoint Inhibitors/administration & dosage
- Immune Checkpoint Inhibitors/adverse effects
Collapse
Affiliation(s)
- Domenico Mallardo
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori - IRCCS - Fondazione 'G. Pascale', Naples, Italy
| | - Francesca Sparano
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori - IRCCS - Fondazione 'G. Pascale', Naples, Italy
| | - Maria Grazia Vitale
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori - IRCCS - Fondazione 'G. Pascale', Naples, Italy
| | - Claudia Trojaniello
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori - IRCCS - Fondazione 'G. Pascale', Naples, Italy
| | - Mario Fordellone
- Universitiy of Campania "Luigi Vanvitelli", 81100, Naples, Italy
| | - Eleonora Cioli
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori - IRCCS - Fondazione 'G. Pascale', Naples, Italy
| | - Assunta Esposito
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori - IRCCS - Fondazione 'G. Pascale', Naples, Italy
| | - Lucia Festino
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori - IRCCS - Fondazione 'G. Pascale', Naples, Italy
| | - Mario Mallardo
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori - IRCCS - Fondazione 'G. Pascale', Naples, Italy
| | - Vito Vanella
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori - IRCCS - Fondazione 'G. Pascale', Naples, Italy
| | - Bianca Arianna Facchini
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori - IRCCS - Fondazione 'G. Pascale', Naples, Italy
| | - Rosaria De Filippi
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Paolo Meinardi
- Division of Surgery of Melanoma and Skin Cancer, Istituto Nazionale Tumori 'Fondazione Pascale' IRCCS, Naples, Italy
| | - Margaret Ottaviano
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori - IRCCS - Fondazione 'G. Pascale', Naples, Italy
| | - Corrado Caracò
- Division of Surgery of Melanoma and Skin Cancer, Istituto Nazionale Tumori 'Fondazione Pascale' IRCCS, Naples, Italy
| | - Ester Simeone
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori - IRCCS - Fondazione 'G. Pascale', Naples, Italy
| | - Paolo Antonio Ascierto
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori - IRCCS - Fondazione 'G. Pascale', Naples, Italy.
| |
Collapse
|
222
|
He Z, Lyu J, Lyu L, Long X, Xu B. Identification of a metabolism-linked genomic signature for prognosis and immunotherapeutic efficiency in metastatic skin cutaneous melanoma. Medicine (Baltimore) 2024; 103:e38347. [PMID: 38847706 PMCID: PMC11155616 DOI: 10.1097/md.0000000000038347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 05/03/2024] [Indexed: 06/10/2024] Open
Abstract
Metastatic skin cutaneous melanoma (MSCM) is the most rapidly progressing/invasive skin-based malignancy, with median survival rates of about 12 months. It appears that metabolic disorders accelerate disease progression. However, correlations between metabolism-linked genes (MRGs) and prognosis in MSCM are unclear, and potential mechanisms explaining the correlation are unknown. The Cancer Genome Atlas (TCGA) was utilized as a training set to develop a genomic signature based on the differentially expressed MRGs (DE-MRGs) between primary skin cutaneous melanoma (PSCM) and MSCM. The Gene Expression Omnibus (GEO) was utilized as a validation set to verify the effectiveness of genomic signature. In addition, a nomogram was established to predict overall survival based on genomic signature and other clinic-based characteristics. Moreover, this study investigated the correlations between genomic signature and tumor micro-environment (TME). This study established a genomic signature consisting of 3 genes (CD38, DHRS3, and TYRP1) and classified MSCM patients into low and high-risk cohorts based on the median risk scores of MSCM cases. It was discovered that cases in the high-risk cohort had significantly lower survival than cases in the low-risk cohort across all sets. Furthermore, a nomogram containing this genomic signature and clinic-based parameters was developed and demonstrated high efficiency in predicting MSCM case survival times. Interestingly, Gene Set Variation Analysis results indicated that the genomic signature was involved in immune-related physiological processes. In addition, this study discovered that risk scoring was negatively correlated with immune-based cellular infiltrations in the TME and critical immune-based checkpoint expression profiles, indicating that favorable prognosis may be influenced in part by immunologically protective micro-environments. A novel 3-genomic signature was found to be reliable for predicting MSCM outcomes and may facilitate personalized immunotherapy.
Collapse
Affiliation(s)
- Zhongshun He
- Department of Oral and Maxillofacial Surgery, Kunming Medical University School and Hospital of Stomatology, Kunming, China
- Yunnan Key Laboratory of Stomatology, Kunming, China
| | - Jing Lyu
- Department of Physiology, Kunming Medical University, Kunming, Yunnan, China
| | - Lechun Lyu
- Technology Transfer Center, Kunming Medical University, Kunming, Yunnan, China
| | - Xiaolin Long
- Yunnan Bestai Biotechnology Co., Ltd., Kunming, Yunnan, China
| | - Biao Xu
- Department of Oral and Maxillofacial Surgery, Kunming Medical University School and Hospital of Stomatology, Kunming, China
- Yunnan Key Laboratory of Stomatology, Kunming, China
| |
Collapse
|
223
|
Yu J, Yu X, Sun X, Wang Q, Long S, Ren R, Guan Z, Yang Z. Bis-2'-F-cG SA SMP isomers encapsulated in cytidinyl/cationic lipids act as potent in situ autologous tumor vaccines. Mol Ther 2024; 32:1917-1933. [PMID: 38637990 PMCID: PMC11184333 DOI: 10.1016/j.ymthe.2024.04.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 10/23/2023] [Accepted: 04/12/2024] [Indexed: 04/20/2024] Open
Abstract
Cancer immunotherapy has greatly improved the prognosis of tumor-bearing patients. Nevertheless, cancer patients exhibit low response rates to current immunotherapy drugs, such as PD1 and PDL1 antibodies. Cyclic dinucleotide analogs are a promising class of immunotherapeutic agents. In this study, in situ autologous tumor vaccines, composed of bis-2'-F-cGSASMP phosphonothioate isomers (FGA-di-pS-2 or FGA-di-pS-4) and cytidinyl/cationic lipids (Mix), were constructed. Intravenous and intratumoral injection of FGA-di-pS-2/Mix or FGA-di-pS-4/Mix enhanced the immunogenic cell death of tumor cells in vivo, leading to the exposure and presentation of whole tumor antigens, inhibiting tumor growth in both LLC and EO771 tumor in situ murine models and increasing their survival rates to 50% and 23%, respectively. Furthermore, the tumor-bearing mice after treatment showed potent immune memory efficacy and exhibited 100% protection against tumor rechallenge. Intravenous administration of FGA-di-pS-2/Mix potently promoted DC maturation, M1 macrophage polarization and CD8+ T cell activation and decreased the proportion of Treg cells in the tumor microenvironment. Notably, two doses of ICD-debris (generated by FGA-di-pS-2 or 4/Mix-treated LLC cells) protected 100% of mice from tumor growth. These tumor vaccines showed promising results and may serve as personalized cancer vaccinations in the future.
Collapse
Affiliation(s)
- Jing Yu
- State Key Laboratory of Natural & Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, No.38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Xiaotong Yu
- State Key Laboratory of Natural & Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, No.38 Xueyuan Road, Haidian District, Beijing 100191, China; Department of Immunology, School of Basic Medical Sciences, Key Laboratory of Medical Immunology of Ministry of Health, Peking University, Beijing 100191, China
| | - Xudong Sun
- State Key Laboratory of Natural & Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, No.38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Quanxin Wang
- State Key Laboratory of Natural & Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, No.38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Sijie Long
- State Key Laboratory of Natural & Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, No.38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Runan Ren
- State Key Laboratory of Natural & Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, No.38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Zhu Guan
- State Key Laboratory of Natural & Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, No.38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Zhenjun Yang
- State Key Laboratory of Natural & Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, No.38 Xueyuan Road, Haidian District, Beijing 100191, China.
| |
Collapse
|
224
|
Liang Y, Xu H, Liu F, Li L, Lin C, Zhang Y, Wang N, Wang L. Immune-related adverse events and their effects on survival outcomes in patients with non-small cell lung cancer treated with immune checkpoint inhibitors: a systematic review and meta-analysis. Front Oncol 2024; 14:1281645. [PMID: 38887231 PMCID: PMC11180722 DOI: 10.3389/fonc.2024.1281645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 05/14/2024] [Indexed: 06/20/2024] Open
Abstract
Background The use of immune checkpoint inhibitors (ICIs) has become the standard of care for non-small cell lung cancer. The purpose of this study was to systematically review the literature to determine whether the occurrence of immune-related adverse events (irAEs) following the use of ICIs predicts different clinical outcomes in non-small cell lung cancer (NSCLC). Methods Relevant studies from the time of database creation to July 20, 2023, were systematically searched to explore the differences in clinical outcomes in patients with advanced NSCLC with or without irAEs. The outcome indicators included the occurrence of irAEs, progression-free survival (PFS), and overall survival (OS). Results 25 studies met the inclusion criteria. Of these studies, 22 reported the effect on OS, and 19 reported the effect on PFS. The results showed that for patients with NSCLC, the occurrence of irAEs after receiving immunotherapy showed a statistically significant benefit over the absence of irAEs for OS (HR=0.55,95% CI=0.46-0.65) and PFS (HR=0.55 95% CI=0.48-0.64), but severe irAEs (grades 3-5) were associated with worse OS (HR=1.05, 95% CI=0.87-1.27). Compared with gastrointestinal, lung, and hepatitis, irAEs of the skin and endocrine system tend to predict better OS and PFS. Conclusion The occurrence of irAEs, especially mild and early irAEs, indicates better OS and PFS in patients with NSCLC treated with ICIs, irrespective of patient characteristics, type of ICIs, and irAEs. However, Grade 3 or higher toxicities resulted in worse OS. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42023409444.
Collapse
Affiliation(s)
- Yuxiang Liang
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Haidi Xu
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Futao Liu
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lei Li
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - ChenXi Lin
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yaozhong Zhang
- Department of Infection, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Na Wang
- Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lei Wang
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
225
|
Piffoux M, Ozenne B, De Backer M, Buyse M, Chiem JC, Péron J. Restricted Net Treatment Benefit in oncology. J Clin Epidemiol 2024; 170:111340. [PMID: 38570079 DOI: 10.1016/j.jclinepi.2024.111340] [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: 11/17/2023] [Revised: 02/11/2024] [Accepted: 03/25/2024] [Indexed: 04/05/2024]
Abstract
OBJECTIVES The restricted Net Treatment Benefit (rNTB) is a clinically meaningful and tractable estimand of the overall treatment effect assessed in randomized trials when at least one survival endpoint with time restriction is used. Its interpretation does not rely on parametric assumptions such as proportional hazards, can be estimated without bias even in the presence of independent right-censoring, and can include a prespecified threshold of minimal clinically relevant difference. To demonstrate that the rNTB, corresponding to the NTB during a predefined time interval, is a meaningful and adaptable measure of treatment effect in clinical trials. METHODS In this simulation study, we tested the impact on the rNTB value, estimation, and power of several factors including the presence of a delayed treatment effect, minimal clinically relevant difference threshold value, restriction time value, and the inclusion of both efficacy and toxicity in the rNTB definition. The impact of right censoring on rNTB was assessed in terms of bias. rNTB-derived statistical tests and log rank (LR) tests were compared in terms of power. RESULTS RNTB estimates are unbiased even in case of right-censoring. rNTB may be used to estimate the benefit/risk ratio of a new treatment, for example, taking into account both survival and toxicity and include several prioritized outcomes. The estimated rNTB is much easier to interpret in this context compared to NTB in the presence of censoring since the latter is intrinsically dependent on the follow-up duration. Including toxicity increases the test power when the experimental treatment is less toxic. rNTB-derived test power increases when the experimental treatment is associated with longer survival and lower toxicity and might increase in the presence of a cure rate or a delayed treatment effect. Case applications on the PRODIGE, Checkmate-066, and Checkmate-067 trials are provided. CONCLUSIONS RNTB is an interesting alternative to describe and test the treatment's effect in a clear and understandable way in case of restriction, particularly in scenarios with nonproportional hazards or when trying to balance benefit and safety. It can be tuned to take into consideration short- or long-term survival differences and one or more prioritized outcomes.
Collapse
Affiliation(s)
- Max Piffoux
- Medical Oncology, Hospices Civils de Lyon, CITOHL, Lyon, France; Direction de la Recherche Clinique et de l'Innovation, Centre Léon Bérard, Lyon, France; Laboratoire MSC Matière et Systèmes Complexes, Université de Paris, CNRS UMR 7057, 75006 Paris, France.
| | - Brice Ozenne
- Neurobiology Research Unit and BrainDrugs, Copenhagen University Hospital, Rigshospitalet, 6-8 Inge Lehmanns Vej, 2100 Copenhagen, Denmark; Section of Biostatistics, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Mickaël De Backer
- International Drug Development Institute (IDDI), Louvain-la-Neuve, Belgium
| | - Marc Buyse
- International Drug Development Institute (IDDI), Louvain-la-Neuve, Belgium; I-BioStat, University of Hasselt, Hasselt, Belgium
| | | | - Julien Péron
- Hospices Civils de Lyon, Oncology Department, Pierre-Bénite, France; Université de Lyon, Université Lyon 1, Reshape Laboratory INSERM U1290, Lyon, France
| |
Collapse
|
226
|
Li KH, Cheung PCF, Petrella TM, Zhang L, Poon IDT, Menjak IB. Clinical outcomes of multisite moderate to high dose radiotherapy for patients with metastatic melanoma. PRECISION RADIATION ONCOLOGY 2024; 8:62-69. [PMID: 40336647 PMCID: PMC11934978 DOI: 10.1002/pro6.1224] [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: 10/11/2023] [Revised: 01/24/2024] [Accepted: 02/22/2024] [Indexed: 05/09/2025] Open
Abstract
Background and purpose This study aimed to summarize the clinical outcomes of patients with metastatic melanoma who received high-dose radiation prior to or during systemic therapy at a single academic institution. Methods We identified patients with metastatic melanoma who underwent high-dose radiation therapy (HDRT) for extracranial metastases prior to or during systemic therapy from 2010 to 2018. Treatment indications included oligometastases, oligoprogression, and local control. Using the Kaplan-Meier method, we plotted overall survival (OS), progression-free survival-1 (PFS1), and PFS2. Competing risk analysis determined the cumulative incidence of local failure (LF) and the time to start or change systemic therapy (SCST). Univariate and multivariable analyses were used to identify predictive factors. Results We analyzed 34 patients with 79 lesions, with a median follow-up of 17.4 months. Sixty-eight percent of patients received systemic therapy after the first HDRT. The median OS was 22 months, with brain metastases before HDRT being a significant predictor in multivariable analysis. The median PFS1 for first-line HDRT was 4.1 months, and the median PFS2 was 3.9 months. Rates of LF were 10.3% at 12 months and 11.7% at 24 months. The incidence of SCST following HDRT was 59.8% at 12 months and 76.1% at 24 months, with radiation targeted at the lung associated with a lower incidence of SCST. Conclusion HDRT for treating metastatic lesions in melanoma demonstrated excellent local control and may play a role in delaying SCST. Additional courses of HDRT may provide cumulative benefits.
Collapse
Affiliation(s)
- Kelly H. Li
- Department of Medical OncologyBC Cancer AgencyVancouverCanada
| | - Patrick CF. Cheung
- Department of Radiation OncologyOdette Cancer Centre, Toronto, Sunnybrook Health Sciences CentreTorontoCanada
| | - Teresa M. Petrella
- Department of Medical OncologyOdette Cancer Centre, Sunnybrook Health Sciences CentreTorontoCanada
| | | | - Ian DT. Poon
- Department of Radiation OncologyOdette Cancer Centre, Toronto, Sunnybrook Health Sciences CentreTorontoCanada
| | - Ines B. Menjak
- Department of Medical OncologyOdette Cancer Centre, Sunnybrook Health Sciences CentreTorontoCanada
| |
Collapse
|
227
|
Oya K, Nakamura Y, Shen LTW, Ishizuki S, Matsusaka S, Fujisawa Y. Soluble PD-L1 predicts tumor response and immune-related adverse events in patients with advanced melanoma treated with anti-PD-1 antibodies. J Dermatol 2024; 51:807-815. [PMID: 38433350 DOI: 10.1111/1346-8138.17183] [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: 12/01/2023] [Revised: 02/14/2024] [Accepted: 02/19/2024] [Indexed: 03/05/2024]
Abstract
Immune checkpoint inhibitors (ICIs) bring prognostic benefits to patients with malignancies. However, there is a substantial number of patients whose lesions are not improved by ICIs. In addition, ICIs may cause immune-related adverse events (irAEs), which could lead to an unfavorable prognosis with fatal consequences. Therefore, we conducted a retrospective study to evaluate the utility of circulating sPD-L1 (soluble programmed cell death 1 ligand 1) as a biomarker in patients with advanced melanoma treated with anti-PD-1 (programmed cell death 1 protein) antibodies. Sera from 31 consecutive patients were prospectively collected before and after anti-PD-1 antibody treatment and the serum level of sPD-L1 was evaluated. We found that high sPD-L1 levels before treatment were associated with better prognosis, and this association was observed only in patients with a low tumor burden. We also found that sPD-L1 levels were elevated in patients who developed severe irAEs after treatment, and the patients with severe irAEs had significantly higher fluctuations in sPD-L1 (delta sPD-L1) than those without severe irAEs. Our study suggests that serum sPD-L1 level is a useful biomarker to predict tumor response and irAE development in patients with advanced melanoma treated with anti-PD-1 antibodies.
Collapse
Affiliation(s)
- Kazumasa Oya
- Department of Dermatology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yoshiyuki Nakamura
- Department of Dermatology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Larina Tzu-Wei Shen
- Department of Clinical Research and Regional Innovation, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Shoichiro Ishizuki
- Department of Dermatology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Satoshi Matsusaka
- Department of Clinical Research and Regional Innovation, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
- Tsukuba Clinical Research and Development Organization, University of Tsukuba, Tsukuba, Japan
| | - Yasuhiro Fujisawa
- Department of Dermatology, Ehime University Graduate School of Medicine, Toon, Japan
| |
Collapse
|
228
|
Oh EL, Dias P, Al-Ogaili Z, Otto J, Warburton L. Unresectable cardiac metastasis from melanoma responds well to combination immunotherapy-a case report. Eur Heart J Case Rep 2024; 8:ytae262. [PMID: 38868157 PMCID: PMC11167960 DOI: 10.1093/ehjcr/ytae262] [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: 09/30/2023] [Revised: 05/09/2024] [Accepted: 05/23/2024] [Indexed: 06/14/2024]
Abstract
Background Melanoma can metastasize to distal organs including the heart although presentation with a symptomatic cardiac metastasis is rare. The optimal management remains uncertain particularly in the era of immunotherapy. Case summary We report a case presenting with a large unresectable cardiac metastasis from melanoma that responded well to treatment with immunotherapy. Conclusion Melanoma can metastasize to the heart and is often challenging to diagnose. Combination immunotherapy can be an effective treatment option even in the setting of a symptomatic and unresectable cardiac metastasis.
Collapse
Affiliation(s)
- Ek Leone Oh
- Medical Oncology Department, Fiona Stanley Hospital, 11 Robin Warren Drive, Murdoch, WA 6150, Australia
| | - Peter Dias
- Cardiology Department, Fiona Stanley Hospital, 11 Robin Warren Drive, Murdoch, WA 6150, Australia
- Advara Heartcare, Wexford Medical Centre, Murdoch, WA 6150, Australia
| | - Zeyad Al-Ogaili
- Nuclear Medicine Department, Fiona Stanley Hospital, 11 Robin Warren Drive, Murdoch, WA 6150, Australia
| | - Jacobus Otto
- Radiology Department, Fiona Stanley Hospital, 11 Robin Warren Drive, Murdoch, WA 6150, Australia
| | - Lydia Warburton
- Medical Oncology Department, Fiona Stanley Hospital, 11 Robin Warren Drive, Murdoch, WA 6150, Australia
- Centre for Precision Health, Edith Cowan University, Joondalup, WA 6027, Australia
| |
Collapse
|
229
|
Langguth M, Maranou E, Koskela SA, Elenius O, Kallionpää RE, Birkman EM, Pulkkinen OI, Sundvall M, Salmi M, Figueiredo CR. TIMP-1 is an activator of MHC-I expression in myeloid dendritic cells with implications for tumor immunogenicity. Genes Immun 2024; 25:188-200. [PMID: 38777826 PMCID: PMC11178497 DOI: 10.1038/s41435-024-00274-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 04/11/2024] [Accepted: 04/19/2024] [Indexed: 05/25/2024]
Abstract
Immune checkpoint therapies (ICT) for advanced solid tumors mark a new milestone in cancer therapy. Yet their efficacy is often limited by poor immunogenicity, attributed to inadequate priming and generation of antitumor T cells by dendritic cells (DCs). Identifying biomarkers to enhance DC functions in such tumors is thus crucial. Tissue Inhibitor of Metalloproteinases-1 (TIMP-1), recognized for its influence on immune cells, has an underexplored relationship with DCs. Our research reveals a correlation between high TIMP1 levels in metastatic melanoma and increased CD8 + T cell infiltration and survival. Network studies indicate a functional connection with HLA genes. Spatial transcriptomic analysis of a national melanoma cohort revealed that TIMP1 expression in immune compartments associates with an HLA-A/MHC-I peptide loading signature in lymph nodes. Primary human and bone-marrow-derived DCs secrete TIMP-1, which notably increases MHC-I expression in classical type 1 dendritic cells (cDC1), especially under melanoma antigen exposure. TIMP-1 affects the immunoproteasome/TAP complex, as seen by upregulated PSMB8 and TAP-1 levels of myeloid DCs. This study uncovers the role of TIMP-1 in DC-mediated immunogenicity with insights into CD8 + T cell activation, providing a foundation for mechanistic exploration and highlighting its potential as a new target for combinatorial immunotherapy to enhance ICT effectiveness.
Collapse
Affiliation(s)
- Miriam Langguth
- Medical Immune Oncology Research Group (MIORG), Institute of Biomedicine, Faculty of Medicine, University of Turku, Turku, Finland
| | - Eleftheria Maranou
- Medical Immune Oncology Research Group (MIORG), Institute of Biomedicine, Faculty of Medicine, University of Turku, Turku, Finland
| | - Saara A Koskela
- Medical Immune Oncology Research Group (MIORG), Institute of Biomedicine, Faculty of Medicine, University of Turku, Turku, Finland
| | - Oskar Elenius
- Medical Immune Oncology Research Group (MIORG), Institute of Biomedicine, Faculty of Medicine, University of Turku, Turku, Finland
| | - Roosa E Kallionpää
- Auria Biobank, University of Turku and Turku University Hospital, Turku, Finland
| | - Eva-Maria Birkman
- Department of Pathology, Laboratory Division, Turku University Hospital and University of Turku, Kiinamyllynkatu 10, 20520, Turku, Finland
| | - Otto I Pulkkinen
- Medical Immune Oncology Research Group (MIORG), Institute of Biomedicine, Faculty of Medicine, University of Turku, Turku, Finland
| | - Maria Sundvall
- Cancer Research Unit, Institute of Biomedicine, and FICAN West Cancer Center Laboratory, University of Turku, and Turku University Hospital, Kiinamyllynkatu 10, 20520, Turku, Finland
- Department of Oncology, Turku University Hospital, Turku, Finland
| | - Marko Salmi
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
- Institute of Biomedicine, University of Turku, Turku, Finland
- MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Carlos R Figueiredo
- Medical Immune Oncology Research Group (MIORG), Institute of Biomedicine, Faculty of Medicine, University of Turku, Turku, Finland.
- Cancer Research Unit, Institute of Biomedicine, and FICAN West Cancer Center Laboratory, University of Turku, and Turku University Hospital, Kiinamyllynkatu 10, 20520, Turku, Finland.
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland.
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Tykistökatu 6, 20520, Turku, Finland.
| |
Collapse
|
230
|
Wilson BE, Sengar M, Tregear M, van der Graaf WTA, Luca Battisti NM, Csaba DL, Soto-Perez-de-Celis E, Gyawali B, Booth CM. Common Sense Oncology: Equity, Value, and Outcomes That Matter. Am Soc Clin Oncol Educ Book 2024; 44:e100039. [PMID: 38788178 DOI: 10.1200/edbk_100039] [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: 05/26/2024]
Abstract
While some recent drug treatments have been transformative for patients with cancer, many treatments offer small benefits despite high clinical toxicity, time toxicity and financial toxicity. Moreover, treatments that do provide substantial clinical benefits are not available to many patients globally due to issues with availability and affordability. The Common Sense Oncology's vision is that patients will have access to treatments that provide meaningful improvements in outcomes that matter, regardless of where they live. In recognition of the growing challenges in the field of oncology, Common Sense Oncology seeks to achieve this vision by improving evidence generation, evidence interpretation and evidence communication.
Collapse
Affiliation(s)
- Brooke E Wilson
- Department of Oncology, Queen's University, Kingston, Canada
- Division of Cancer Care and Epidemiology, Queen's Cancer Research Institute, Kingston, Canada
- School of Population Health, Faculty of Medicine, University of New South Wales, Randwick, Australia
| | - Manju Sengar
- Tata Memorial Hospital, Affiliated to Homi Bhabha National Institute, Mumbai, India
| | | | - Winette T A van der Graaf
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Nicolò Matteo Luca Battisti
- Department of Medicine, Breast Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom
- Inequalities Focused Topic Network, European Cancer Organisation, Brussels, Belgium
- International Society of Geriatric Oncology, Geneva, Switzerland
| | - Degi Laszlo Csaba
- Faculty of Sociology and Social Work, Babeş-Bolyai University, Cluj Napoca, Romania
| | - Enrique Soto-Perez-de-Celis
- Department of Geriatrics, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO
| | - Bishal Gyawali
- Department of Oncology, Queen's University, Kingston, Canada
- Division of Cancer Care and Epidemiology, Queen's Cancer Research Institute, Kingston, Canada
| | - Christopher M Booth
- Department of Oncology, Queen's University, Kingston, Canada
- Division of Cancer Care and Epidemiology, Queen's Cancer Research Institute, Kingston, Canada
| |
Collapse
|
231
|
Monnickendam G. Assessing the Performance of Alternative Methods for Estimating Long-Term Survival Benefit of Immuno-oncology Therapies. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2024; 27:746-754. [PMID: 38428815 DOI: 10.1016/j.jval.2024.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 02/18/2024] [Accepted: 02/22/2024] [Indexed: 03/03/2024]
Abstract
OBJECTIVES This study aimed to determine the accuracy and consistency of established methods of extrapolating mean survival for immuno-oncology (IO) therapies, the extent of any systematic biases in estimating long-term clinical benefit, what influences the magnitude of any bias, and the potential implications for health technology assessment. METHODS A targeted literature search was conducted to identify published long-term follow-up from clinical trials of immune-checkpoint inhibitors. Earlier published results were identified and Kaplan-Meier estimates for short- and long-term follow-up were digitized and converted to pseudo-individual patient data using an established algorithm. Six standard parametric, 5 flexible parametric, and 2 mixture-cure models (MCMs) were used to extrapolate long-term survival. Mean and restricted mean survival time (RMST) were estimated and compared between short- and long-term follow-up. RESULTS Predicted RMST from extrapolation of early data underestimated observed RMST in long-term follow-up for 184 of 271 extrapolations. All models except the MCMs frequently underestimated observed RMST. Mean survival estimates increased with longer follow-up in 196 of 270 extrapolations. The increase exceeded 20% in 122 extrapolations. Log-logistic and log-normal models showed the smallest change with additional follow-up. MCM performance varied substantially with functional form. CONCLUSIONS Standard and flexible parametric models frequently underestimate mean survival for IO treatments. Log-logistic and log-normal models may be the most pragmatic and parsimonious solutions for estimating IO mean survival from immature data. Flexible parametric models may be preferred when the data used in health technology assessment are more mature. MCMs fitted to immature data produce unreliable results and are not recommended.
Collapse
|
232
|
Townsend MJ, Benque IJ, Li M, Grover S. Review article: Contemporary management of gastrointestinal, pancreatic and hepatic toxicities of immune checkpoint inhibitors. Aliment Pharmacol Ther 2024; 59:1350-1365. [PMID: 38590108 DOI: 10.1111/apt.17980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/13/2023] [Accepted: 03/21/2024] [Indexed: 04/10/2024]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) are effective oncologic agents which frequently cause immune-related adverse events (irAEs) which can impact multiple organ systems. Onco-Gastroenterology is a novel and emerging subspecialty within gastroenterology focused on cancer treatment-related complications. Gastroenterologists must be prepared to identify and manage diverse immune-mediated toxicities including enterocolitis, hepatitis, pancreatitis and other ICI-induced toxicities. AIM To provide a narrative review of the epidemiology, diagnostic evaluation and management of checkpoint inhibitor-induced gastrointestinal and hepatic toxicities. METHODS We searched Cochrane and PubMed databases for articles published through August 2023. RESULTS Gastrointestinal and hepatic irAEs include most commonly enterocolitis and hepatitis, but also pancreatitis, oesophagitis, gastritis, motility disorders (gastroparesis) and other rarer toxicities. Guidelines from the National Comprehensive Cancer Network, American Society of Clinical Oncology and European Society for Medical Oncology, in combination with emerging cohort and clinical trial data, offer strategies for management of ICI toxicities. Evaluation of irAEs severity by formal classification and clinical stability, and a thorough workup for alternative etiologies which may clinically mimic irAEs underlie initial management. Treatments include corticosteroids, biologics and other immunosuppressive agents plus supportive care; decisions on dosing, timing and choice of steroid adjuncts and potential for subsequent checkpoint inhibitor dosing are nuanced and toxicity-specific. CONCLUSIONS Expanding clinical trial and cohort data have clarified the epidemiology and clinical characteristics of gastrointestinal, pancreatic and hepatic toxicities of ICIs. Guidelines, though valuable, remain based principally on retrospective cohort data. Quality prospective, controlled studies may refine algorithms for treatment and potential immunotherapy rechallenge.
Collapse
Affiliation(s)
- Matthew J Townsend
- Department of Medicine, Duke University Hospital, Durham, North Carolina, USA
| | - Isaac J Benque
- University of California San Francisco School of Medicine, San Francisco, California, USA
| | - Michael Li
- University of California San Francisco School of Medicine, San Francisco, California, USA
- Division of Gastroenterology, University of California San Francisco Medical Center, San Francisco, California, USA
| | - Shilpa Grover
- Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
233
|
Cariou PL, Pobel C, Michot JM, Danlos FX, Besse B, Carbonnel F, Mariette X, Marabelle A, Messayke S, Robert C, Routier E, Noël N, Lambotte O. Impact of immunosuppressive agents on the management of immune-related adverse events of immune checkpoint blockers. Eur J Cancer 2024; 204:114065. [PMID: 38643707 DOI: 10.1016/j.ejca.2024.114065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 04/11/2024] [Accepted: 04/11/2024] [Indexed: 04/23/2024]
Abstract
BACKGROUND Immune checkpoint blockers (ICBs) can induce immune-related adverse events (irAEs) whose management is based on expert opinion and may require the prescription of steroids and/or immunosuppressants (ISs). Recent data suggest that these treatments can reduce the effectiveness of ICBs. OBJECTIVE To investigate the relationship between the use of steroids and/or ISs and overall survival (OS) and progression-free survival (PFS) among ICB-treated patients with an irAE. METHODS We prospectively collected data from the medical records of patients with solid tumors or lymphoma in the French REISAMIC cohort and who had been treated with ICBs between June 2014 and June 2020. RESULTS 184 ICB-treated patients experienced at least one Common Terminology Criteria for Adverse Events grade ≥ 2 irAE. 107 (58.2%) were treated with steroids alone, 20 (10.9%) with steroids plus IS, 57 (31.0%) not received steroids or IS. The median OS was significantly shorter for patients treated with steroids alone (25.2 months [95% confidence interval (CI): 22.3-32.4] than for patients treated without steroids or IS (63 months [95%CI: 40.4-NA]) and those receiving an IS with steroids (53.4 months [95%CI: 47.3-NA]) (p < 0.001). The median PFS was significantly shorter for patients treated with steroids alone (17.0 months [95%CI: 11.7-22.9]) than for patients treated without steroids or IS (33.9 months [95%CI: 18.0-NA]) and those receiving an IS with steroids (41.1 months [95%CI: 26.2-NA]) (p = 0.006). There were no significant intergroup differences in the hospital admission and infection rates. CONCLUSION In a prospective cohort of ICB-treated patients, the use of IS was not associated with worse OS or PFS, contrasting with the use of steroids for the management of irAEs.
Collapse
Affiliation(s)
- Pierre-Louis Cariou
- Université Paris Saclay, AP-HP, Hôpital de Bicêtre, Department of Internal Medicine, UMR 1184, CEA INSERM, FHU CARE, Le Kremlin Bicêtre, France
| | - Cédric Pobel
- Drug Development Department (DITEP), Gustave Roussy, 94805 Villejuif, France
| | - Jean-Marie Michot
- Drug Development Department (DITEP), Gustave Roussy, 94805 Villejuif, France
| | | | - Benjamin Besse
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| | - Franck Carbonnel
- AP-HP, Department of Gastroenterology, University Hospital of Bicêtre, Paris Sud University, FHU CARE, 78 Rue du General Leclerc, 94270 Le Kremlin-Bicetre, France
| | - Xavier Mariette
- Université Paris-Saclay, AP-HP, Hôpital Bicêtre, Rheumatology Department, INSERM UMR 1184, FHU CARE, Paris, Le Kremlin Bicêtre, France
| | - Aurélien Marabelle
- Drug Development Department (DITEP), Gustave Roussy, 94805 Villejuif, France
| | - Sabine Messayke
- Gustave Roussy - Paris-Saclay University, Pharmacovigilance Unit, 94800 Villejuif, France
| | - Caroline Robert
- Dermatology Service, Department of Medicine, Gustave Roussy and Paris-Saclay University, 114 Rue Edouard Vaillant, 94805 Villejuif, France
| | - Emilie Routier
- Dermatology Service, Department of Medicine, Gustave Roussy and Paris-Saclay University, 114 Rue Edouard Vaillant, 94805 Villejuif, France
| | - Nicolas Noël
- Université Paris Saclay, AP-HP, Hôpital de Bicêtre, Department of Internal Medicine, UMR 1184, CEA INSERM, FHU CARE, Le Kremlin Bicêtre, France
| | - Olivier Lambotte
- Université Paris Saclay, AP-HP, Hôpital de Bicêtre, Department of Internal Medicine, UMR 1184, CEA INSERM, FHU CARE, Le Kremlin Bicêtre, France.
| |
Collapse
|
234
|
Sedhom R, Bates-Pappas GE, Feldman J, Elk R, Gupta A, Fisch MJ, Soto-Perez-de-Celis E. Tumor Is Not the Only Target: Ensuring Equitable Person-Centered Supportive Care in the Era of Precision Medicine. Am Soc Clin Oncol Educ Book 2024; 44:e434026. [PMID: 39177644 DOI: 10.1200/edbk_434026] [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: 08/24/2024]
Abstract
Communication in oncology has always been challenging. The new era of precision oncology creates prognostic uncertainty. Still, person-centered care requires attention to people and their care needs. Living with cancer portends an experience that is life-altering, no matter what the outcome. Supporting patients and families through this unique experience requires careful attention, honed skills, an understanding of process and balance measures of innovation, and recognizing that supportive care is a foundational element of cancer medicine, rather than an either-or approach, an and-with approach that emphasizes the regular integration of palliative care (PC), geriatric oncology, and skilled communication.
Collapse
Affiliation(s)
- Ramy Sedhom
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Leonard Davis Institute of Health Economics, University of Pennsylvania, Philadelphia, PA
- Penn Center for Cancer Care Innovation, Abramson Cancer Center, Penn Medicine, Philadelphia, PA
| | - Gleneara E Bates-Pappas
- Department of Psychiatry and Behavioral Sciences, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Ronit Elk
- Center for Palliative and Supportive Care, University of Alabama at Birmingham, Birmingham, AL
- Division of Geriatrics, School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Arjun Gupta
- Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis
| | | | - Enrique Soto-Perez-de-Celis
- Department of Geriatrics, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| |
Collapse
|
235
|
Strike A, Barker SB, McGuire N, Kaur G. Hypercalcemia Secondary to Calcitriol Production From Dual Combination Immunotherapy in Pulmonary Metastatic Melanoma. Cureus 2024; 16:e62379. [PMID: 39006637 PMCID: PMC11246753 DOI: 10.7759/cureus.62379] [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: 06/14/2024] [Indexed: 07/16/2024] Open
Abstract
Nivolumab and ipilimumab are immunotherapy agents recommended for the treatment of metastatic melanoma. A rare adverse effect of these agents is hypercalcemia. The mechanism of immunotherapy-mediated hypercalcemia is thought to be due to ectopic calcitriol production from activated macrophages, similar to sarcoidosis. We present a case of a 76-year-old female with metastatic melanoma who developed severe hypercalcemia after completing a cycle of combined nivolumab and ipilimumab therapy. After other common causes of hypercalcemia in malignancy were ruled out, the decision was made to aggressively treat her hypercalcemia while inpatient and hold immunotherapy at discharge. Since holding immunotherapy, she has not had a repeat occurrence of hypercalcemia. This case stresses the importance of including immunotherapy adverse effects in the differential diagnosis for hypercalcemia in malignancy.
Collapse
Affiliation(s)
- Andrew Strike
- Graduate Medical Education, Northeast Georgia Medical Center Gainesville, Gainesville, USA
| | - Steven B Barker
- Internal Medicine, Northeast Georgia Medical Center Gainesville, Gainesville, USA
| | - Nicole McGuire
- Internal Medicine, Northeast Georgia Medical Center Gainesville, Gainesville, USA
| | - Gurleen Kaur
- Internal Medicine, Northeast Georgia Medical Center Gainesville, Gainesville, USA
| |
Collapse
|
236
|
Widman AJ, Shah M, Frydendahl A, Halmos D, Khamnei CC, Øgaard N, Rajagopalan S, Arora A, Deshpande A, Hooper WF, Quentin J, Bass J, Zhang M, Langanay T, Andersen L, Steinsnyder Z, Liao W, Rasmussen MH, Henriksen TV, Jensen SØ, Nors J, Therkildsen C, Sotelo J, Brand R, Schiffman JS, Shah RH, Cheng AP, Maher C, Spain L, Krause K, Frederick DT, den Brok W, Lohrisch C, Shenkier T, Simmons C, Villa D, Mungall AJ, Moore R, Zaikova E, Cerda V, Kong E, Lai D, Malbari MS, Marton M, Manaa D, Winterkorn L, Gelmon K, Callahan MK, Boland G, Potenski C, Wolchok JD, Saxena A, Turajlic S, Imielinski M, Berger MF, Aparicio S, Altorki NK, Postow MA, Robine N, Andersen CL, Landau DA. Ultrasensitive plasma-based monitoring of tumor burden using machine-learning-guided signal enrichment. Nat Med 2024; 30:1655-1666. [PMID: 38877116 PMCID: PMC7616143 DOI: 10.1038/s41591-024-03040-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 04/30/2024] [Indexed: 06/16/2024]
Abstract
In solid tumor oncology, circulating tumor DNA (ctDNA) is poised to transform care through accurate assessment of minimal residual disease (MRD) and therapeutic response monitoring. To overcome the sparsity of ctDNA fragments in low tumor fraction (TF) settings and increase MRD sensitivity, we previously leveraged genome-wide mutational integration through plasma whole-genome sequencing (WGS). Here we now introduce MRD-EDGE, a machine-learning-guided WGS ctDNA single-nucleotide variant (SNV) and copy-number variant (CNV) detection platform designed to increase signal enrichment. MRD-EDGESNV uses deep learning and a ctDNA-specific feature space to increase SNV signal-to-noise enrichment in WGS by ~300× compared to previous WGS error suppression. MRD-EDGECNV also reduces the degree of aneuploidy needed for ultrasensitive CNV detection through WGS from 1 Gb to 200 Mb, vastly expanding its applicability within solid tumors. We harness the improved performance to identify MRD following surgery in multiple cancer types, track changes in TF in response to neoadjuvant immunotherapy in lung cancer and demonstrate ctDNA shedding in precancerous colorectal adenomas. Finally, the radical signal-to-noise enrichment in MRD-EDGESNV enables plasma-only (non-tumor-informed) disease monitoring in advanced melanoma and lung cancer, yielding clinically informative TF monitoring for patients on immune-checkpoint inhibition.
Collapse
Affiliation(s)
- Adam J Widman
- New York Genome Center, New York, NY, USA.
- Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | | | - Amanda Frydendahl
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Daniel Halmos
- New York Genome Center, New York, NY, USA
- Weill Cornell Medicine, New York, NY, USA
| | - Cole C Khamnei
- New York Genome Center, New York, NY, USA
- Weill Cornell Medicine, New York, NY, USA
| | - Nadia Øgaard
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Srinivas Rajagopalan
- New York Genome Center, New York, NY, USA
- Weill Cornell Medicine, New York, NY, USA
| | - Anushri Arora
- New York Genome Center, New York, NY, USA
- Weill Cornell Medicine, New York, NY, USA
| | - Aditya Deshpande
- New York Genome Center, New York, NY, USA
- Weill Cornell Medicine, New York, NY, USA
| | | | - Jean Quentin
- New York Genome Center, New York, NY, USA
- Weill Cornell Medicine, New York, NY, USA
| | - Jake Bass
- New York Genome Center, New York, NY, USA
- Weill Cornell Medicine, New York, NY, USA
| | - Mingxuan Zhang
- New York Genome Center, New York, NY, USA
- Weill Cornell Medicine, New York, NY, USA
| | - Theophile Langanay
- New York Genome Center, New York, NY, USA
- Weill Cornell Medicine, New York, NY, USA
| | - Laura Andersen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | - Will Liao
- New York Genome Center, New York, NY, USA
| | - Mads Heilskov Rasmussen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Tenna Vesterman Henriksen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Sarah Østrup Jensen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Jesper Nors
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Christina Therkildsen
- Gastro Unit, Copenhagen University Hospital, Amager - Hvidovre Hospital, Hvidovre, Denmark
| | - Jesus Sotelo
- New York Genome Center, New York, NY, USA
- Weill Cornell Medicine, New York, NY, USA
| | - Ryan Brand
- New York Genome Center, New York, NY, USA
- Weill Cornell Medicine, New York, NY, USA
| | - Joshua S Schiffman
- New York Genome Center, New York, NY, USA
- Weill Cornell Medicine, New York, NY, USA
| | - Ronak H Shah
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Colleen Maher
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Lavinia Spain
- Cancer Dynamics Laboratory, The Francis Crick Institute, London, UK
- Renal and Skin Unit, The Royal Marsden NHS Foundation Trust, London, UK
| | - Kate Krause
- Mass General Cancer Center, Massachusetts General Hospital, Boston, MA, USA
| | - Dennie T Frederick
- Mass General Cancer Center, Massachusetts General Hospital, Boston, MA, USA
| | - Wendie den Brok
- Department of Medical Oncology, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Caroline Lohrisch
- Department of Medical Oncology, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Tamara Shenkier
- Department of Medical Oncology, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Christine Simmons
- Department of Medical Oncology, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Diego Villa
- Department of Medical Oncology, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Andrew J Mungall
- Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
| | - Richard Moore
- Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
| | - Elena Zaikova
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Viviana Cerda
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Esther Kong
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Daniel Lai
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, British Columbia, Canada
| | | | | | - Dina Manaa
- New York Genome Center, New York, NY, USA
| | | | - Karen Gelmon
- Department of Medical Oncology, BC Cancer Agency, Vancouver, British Columbia, Canada
| | | | - Genevieve Boland
- Mass General Cancer Center, Massachusetts General Hospital, Boston, MA, USA
| | - Catherine Potenski
- New York Genome Center, New York, NY, USA
- Weill Cornell Medicine, New York, NY, USA
| | - Jedd D Wolchok
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Samra Turajlic
- Cancer Dynamics Laboratory, The Francis Crick Institute, London, UK
- Renal and Skin Unit, The Royal Marsden NHS Foundation Trust, London, UK
| | - Marcin Imielinski
- New York Genome Center, New York, NY, USA
- Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA
| | | | - Sam Aparicio
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Michael A Postow
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medicine, New York, NY, USA
| | | | - Claus Lindbjerg Andersen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Dan A Landau
- New York Genome Center, New York, NY, USA.
- Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
237
|
de Joode K, Mora AR, van Schaik RHN, Zippelius A, van der Veldt A, Gerard CL, Läubli H, Michielin O, von Moos R, Joerger M, Levesque MP, Aeppli S, Mangana J, Mangas C, Trost N, Meyer S, Parvex SL, Mathijssen R, Metaxas Y. Effects of CTLA-4 Single Nucleotide Polymorphisms on Toxicity of Ipilimumab-Containing Regimens in Patients With Advanced Stage Melanoma. J Immunother 2024; 47:190-194. [PMID: 38318726 DOI: 10.1097/cji.0000000000000506] [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: 08/30/2023] [Accepted: 10/16/2023] [Indexed: 02/07/2024]
Abstract
Single nucleotide polymorphisms (SNPs) in the cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) gene, an inhibitor of T-cell priming, are associated with auto and alloimmunity. Studies implied a role for these SNPs as surrogate markers for immunotherapy-outcome in patients with melanoma. However, no predictive SNPs are defined to date. We analyzed different CTLA-4 SNPs in a large multicenter cohort of patients with ipilimumab-treated melanoma and investigated possible correlations with treatment-related outcomes. Archival blood and/or tumor tissue samples were collected from 361 patients with advanced-stage ipilimumab-treated (±nivolumab) in 6 Swiss and Dutch hospitals. Matrix-assisted laser desorption/ionization-time of flight mass spectrometry based DNA genotyping was performed for 10 different CTLA-4 SNPs: 49A>G, CT60G>A, Jo27T>C, Jo30G>A, Jo31G>T, -658C>T, -1722T>C, -1661A>G, 318C>T, and C>T rs1863800. Associations between different allele genotypes and occurrence of grade ≥3 adverse events (AEs) and survival were tested using univariable logistic regressions or Cox proportional hazard models. 262/361 (73%) patients could be analyzed; 65% of those were males, the median age was 58 years, 39% showed a partial or complete response, and 65% had ≥1 AEs. A TT-genotype of -1722T>C SNP was significantly associated with a lower incidence of grade ≥3 AEs ( P = 0.049), whereas the GG-genotype of CT60G>A correlated with a higher incidence of grade ≥3 AEs ( P = 0.026). The TT-genotype of Jo27T>C SNP ( P = 0.056) and GG-genotype of Jo31G>T ( P = 0.046) were associated with overall survival. CTLA-4 SNPs might predict treatment-related outcomes in patients with melanoma receiving ipilimumab. Confirmatory studies are needed to fully exploit those findings as predictive biomarkers for ipilimumab AEs.
Collapse
Affiliation(s)
- Karlijn de Joode
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Alfonso Rojas Mora
- Competence Center of Swiss Group for Clinical Cancer Research (SAKK), Bern, Switzerland
| | - Ron H N van Schaik
- Department of Clinical Chemistry, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Alfred Zippelius
- Department of Biomedicine, Division of Medical Oncology, University Hospital and University of Basel, Basel, Switzerland
| | - Astrid van der Veldt
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Camille Léa Gerard
- Precision Oncology Center, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Heinz Läubli
- Department of Biomedicine, Division of Medical Oncology, University Hospital and University of Basel, Basel, Switzerland
| | - Olivier Michielin
- Department of Medical Oncology, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Roger von Moos
- Department of Oncology/Hematology, Cantonal Hospital Graubünden, Chur, Switzerland
| | - Markus Joerger
- Department of Oncology/Hematology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | | | - Stefanie Aeppli
- Department of Oncology/Hematology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Johanna Mangana
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Cristina Mangas
- Oncology Institute of Southern Switzerland (IOSI), Bellinzona, Switzerland
| | - Nadine Trost
- Department of Molecular Diagnostics and Research, Blood Transfusion Service Zurich, Swiss Red Cross, Schlieren, Switzerland
| | - Stefan Meyer
- Department of Molecular Diagnostics and Research, Blood Transfusion Service Zurich, Swiss Red Cross, Schlieren, Switzerland
| | | | - Ron Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Yannis Metaxas
- Department of Medical Oncology, Cantonal Hospital Muensterlingen, Muensterlingen, Switzerland
| |
Collapse
|
238
|
Mountzios G, Naidoo J, Wang C, Creelan BC, Trotier DC, Campbell TC, Peters S. Beyond Chemoimmunotherapy in Advanced Non-Small Cell Lung Cancer: New Frontiers, New Challenges. Am Soc Clin Oncol Educ Book 2024; 44:e432526. [PMID: 38781566 DOI: 10.1200/edbk_432526] [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
Chemoimmunotherapy is currently the preferred first-line treatment option for the majority of patients with advanced non-small cell lung cancer without driver genetic alterations. Most of these patients, however, will experience disease progression within the first year after treatment initiation and both patients and their physicians will be confronted with the dilemma of the optimal second-line treatment. Identification of molecular targets, such as KRASG12C, BRAFV600X, METexon14, and human epidermal growth factor receptor 2 mutations, and RET rearrangements offer therapeutic opportunities in pretreated patients with corresponding alterations. For those tumors that do not harbor oncogenic drivers, second-line treatment with docetaxel remains the current standard of care despite modest efficacy. Strategies to challenge docetaxel include the combination of immune checkpoint inhibitors (ICIs) with tyrosine inhibitors of multiple kinases or with DNA damage response inhibitors, antibody-drug conjugates, and locoregional treatments for oligoprogressive disease. Next-generation immunotherapy strategies, such as T-cell engagers, immune-mobilizing monoclonal T-cell receptors, chimeric antigen receptor cell therapy, tumor infiltrating lymphocytes, and T-cell receptor cell therapy are being currently investigated in the quest to reverse resistance to ICIs. Importantly, the advent of these new agents heralds a novel spectrum of toxicities that require both the physician's and the patient's education. Herein, we review current and future strategies aiming to outperform docetaxel after chemoimmunotherapy failure, and we provide practical information on how to best communicate to our patients the unique toxicity aspects associated with immunotherapy.
Collapse
Affiliation(s)
- Giannis Mountzios
- 4th Department of Medical Oncology and Clinical Trials Unit, Henry Dunant Hospital Center, Athens, Greece
| | - Jarushka Naidoo
- Department of Oncology, Beaumont Hospital, Beaumont RCSI Cancer Centre, Dublin, Ireland
- RCSI University of Health Sciences, Dublin, Ireland
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD
| | - Chao Wang
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Benjamin C Creelan
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Daniel C Trotier
- Department of Hematology-Oncology, University of Wisconsin-Madison, Madison, WI
| | - Toby C Campbell
- Department of Hematology-Oncology, University of Wisconsin-Madison, Madison, WI
| | - Solange Peters
- Oncology Department, CHUV, Lausanne University, Lausanne, Switzerland
| |
Collapse
|
239
|
Proulx-Rocray F, Soulières D. Emerging monoclonal antibody therapy for head and neck squamous cell carcinoma. Expert Opin Emerg Drugs 2024; 29:165-176. [PMID: 38616696 DOI: 10.1080/14728214.2024.2339906] [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: 12/21/2023] [Accepted: 04/03/2024] [Indexed: 04/16/2024]
Abstract
INTRODUCTION The incidence of head and neck squamous cell carcinoma (HNSCC) is increasing, particularly among younger populations. It is projected that the number of new cases will increase by almost 50% by 2040, with market revenues expected to triple in the same period. Despite the recent introduction of immune checkpoint inhibitors (ICIs) into the therapeutic armamentarium, the vast majority of patients with recurrent and/or metastatic (R/M) HNSCC fail to derive durable benefits from systemic therapy. AREAS COVERED This article aims to review the multiple monoclonal antibodies (mAbs) regimens currently under development, targeting various growth factors, immune checkpoints, immune costimulatory receptors, and more. EXPERT OPINION So far, the combination of anti-EGFR and ICI appears to be the most promising, especially in HPV-negative patients. It will be interesting to confirm whether the arrival of antibody-drug conjugates and bispecific mAb can surpass the efficacy of anti-EGFR, as they are also being tested in combination with ICI. Furthermore, we believe that immune costimulatory agonists and various ICIs combination are worth monitoring, despite some initial setbacks.
Collapse
Affiliation(s)
- Francis Proulx-Rocray
- Hematology and Medical Oncology Department, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
| | - Denis Soulières
- Hematology and Medical Oncology Department, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
| |
Collapse
|
240
|
Halpert MM, Burns BA, Rosario SR, Withers HG, Trivedi AJ, Hofferek CJ, Gephart BD, Wang H, Vazquez-Perez J, Amanya SB, Hyslop ST, Yang J, Kemnade JO, Sandulache VC, Konduri V, Decker WK. Multifactoral immune modulation potentiates durable remission in multiple models of aggressive malignancy. FASEB J 2024; 38:e23644. [PMID: 38738472 PMCID: PMC11155525 DOI: 10.1096/fj.202302675r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 04/05/2024] [Accepted: 04/22/2024] [Indexed: 05/14/2024]
Abstract
Tumors typically lack canonical danger signals required to activate adaptive immunity and also frequently employ substantial immunomodulatory mechanisms that downregulate adaptive responses and contribute to escape from immune surveillance. Given the variety of mechanisms involved in shielding tumors from immune recognition, it is not surprising that single-agent immunomodulatory approaches have been largely unsuccessful in generating durable antitumor responses. Here we report a unique combination of immunomodulatory and cytostatic agents that recondition the tumor microenvironment and eliminate complex and/or poor-prognosis tumor types including the non-immunogenic 4T-1 model of TNBC, the aggressive MOC-2 model of HNSCC, and the high-risk MYCN-amplified model of neuroblastoma. A course of therapy optimized for TNBC cured a majority of tumors in both ectopic and orthotopic settings and eliminated metastatic spread in all animals tested at the highest doses. Immune responses were transferable between therapeutic donor and naïve recipient through adoptive transfer, and a sizeable abscopal effect on distant, untreated lesions could be demonstrated experimentally. Similar results were observed in HNSCC and neuroblastoma models, with characteristic remodeling of the tumor microenvironment documented in all model systems. scRNA-seq analysis implicated upregulation of innate immune responses and antigen presentation in tumor cells and the myeloid cell compartment as critical early events. This analysis also highlighted the potential importance of the autonomic nervous system in the governance of inflammatory processes. The data indicate that the targeting of multiple pathways and mechanisms of action can result in substantial synergistic antitumor effects and suggest follow-up in the neoadjuvant setting may be warranted.
Collapse
Affiliation(s)
- MM Halpert
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030 United States
| | - BA Burns
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030 United States
| | - SR Rosario
- Department of Biostatistics and Bioinformatics, Baylor College of Medicine, Houston, TX 77030 United States
- Acquired Resistance to Therapy Network (ARTNet) U24/U54 Investigator, Baylor College of Medicine, Houston, TX 77030 United States
| | - HG Withers
- Department of Biostatistics and Bioinformatics, Baylor College of Medicine, Houston, TX 77030 United States
| | - AJ Trivedi
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030 United States
| | - CJ Hofferek
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030 United States
| | - BD Gephart
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030 United States
| | - H Wang
- Department of Medicine, Section of Hematology & Oncology, Baylor College of Medicine, Houston, TX 77030 United States
| | - J Vazquez-Perez
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030 United States
| | - SB Amanya
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030 United States
| | - ST Hyslop
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030 United States
| | - J Yang
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030 United States
| | - JO Kemnade
- Department of Medicine, Section of Hematology & Oncology, Baylor College of Medicine, Houston, TX 77030 United States
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030 United States
| | - VC Sandulache
- Acquired Resistance to Therapy Network (ARTNet) U24/U54 Investigator, Baylor College of Medicine, Houston, TX 77030 United States
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030 United States
- Bobby R. Alford Department of Otolaryngology - Head and Neck Surgery, Baylor College of Medicine, Houston, TX 77030 United States
| | - V Konduri
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030 United States
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030 United States
| | - WK Decker
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030 United States
- Acquired Resistance to Therapy Network (ARTNet) U24/U54 Investigator, Baylor College of Medicine, Houston, TX 77030 United States
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030 United States
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030 United States
| |
Collapse
|
241
|
Gregucci F, Beal K, Knisely JPS, Pagnini P, Fiorentino A, Bonzano E, Vanpouille-Box CI, Cisse B, Pannullo SC, Stieg PE, Formenti SC. Biological Insights and Radiation-Immuno-Oncology Developments in Primary and Secondary Brain Tumors. Cancers (Basel) 2024; 16:2047. [PMID: 38893165 PMCID: PMC11171192 DOI: 10.3390/cancers16112047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Malignant central nervous system (CNS) cancers include a group of heterogeneous dis-eases characterized by a relative resistance to treatments and distinguished as either primary tumors arising in the CNS or secondary tumors that spread from other organs into the brain. Despite therapeutic efforts, they often cause significant mortality and morbidity across all ages. Radiotherapy (RT) remains the main treatment for brain cancers, improving associated symptoms, improving tumor control, and inducing a cure in some. However, the ultimate goal of cancer treatment, to improve a patient's survival, remains elusive for many CNS cancers, especially primary tumors. Over the years, there have thus been many preclinical studies and clinical trials designed to identify and overcome mechanisms of resistance to improve outcomes after RT and other therapies. For example, immunotherapy delivered concurrent with RT, especially hypo-fractionated stereotactic RT, is synergistic and has revolutionized the clinical management and outcome of some brain tumors, in particular brain metastases (secondary brain tumors). However, its impact on gliomas, the most common primary malignant CNS tumors, remains limited. In this review, we provide an overview of radioresistance mechanisms, the emerging strategies to overcome radioresistance, the role of the tumor microenviroment (TME), and the selection of the most significant results of radiation-immuno-oncological investigations. We also identify novel therapeutic opportunities in primary and secondary brain tumors with the purpose of elucidating current knowledge and stimulating further research to improve tumor control and patients' survival.
Collapse
Affiliation(s)
- Fabiana Gregucci
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA; (F.G.); (K.B.); (J.P.S.K.); (P.P.); (C.I.V.-B.)
- Department of Radiation Oncology, Miulli General Regional Hospital, Acquaviva delle Fonti, 70021 Bari, Italy;
| | - Kathryn Beal
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA; (F.G.); (K.B.); (J.P.S.K.); (P.P.); (C.I.V.-B.)
| | - Jonathan P. S. Knisely
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA; (F.G.); (K.B.); (J.P.S.K.); (P.P.); (C.I.V.-B.)
| | - Paul Pagnini
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA; (F.G.); (K.B.); (J.P.S.K.); (P.P.); (C.I.V.-B.)
| | - Alba Fiorentino
- Department of Radiation Oncology, Miulli General Regional Hospital, Acquaviva delle Fonti, 70021 Bari, Italy;
- Department of Medicine and Surgery, LUM University, Casamassima, 70010 Bari, Italy
| | - Elisabetta Bonzano
- Department of Radiation Oncology, IRCCS San Matteo Polyclinic Foundation, 27100 Pavia, Italy;
| | - Claire I. Vanpouille-Box
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA; (F.G.); (K.B.); (J.P.S.K.); (P.P.); (C.I.V.-B.)
- Sandra and Edward Meyer Cancer Center, New York, NY 10065, USA
| | - Babacar Cisse
- Department of Neurological Surgery, Weill Cornell Medicine, New York, NY 10065, USA; (B.C.); (S.C.P.); (P.E.S.)
| | - Susan C. Pannullo
- Department of Neurological Surgery, Weill Cornell Medicine, New York, NY 10065, USA; (B.C.); (S.C.P.); (P.E.S.)
- Department of Biomedical Engineering, College of Engineering, Cornell University, Ithaca, NY 14850, USA
| | - Philip E. Stieg
- Department of Neurological Surgery, Weill Cornell Medicine, New York, NY 10065, USA; (B.C.); (S.C.P.); (P.E.S.)
- Department of Radiology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Silvia C. Formenti
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA; (F.G.); (K.B.); (J.P.S.K.); (P.P.); (C.I.V.-B.)
- Sandra and Edward Meyer Cancer Center, New York, NY 10065, USA
| |
Collapse
|
242
|
Shalata W, Attal ZG, Solomon A, Shalata S, Abu Saleh O, Tourkey L, Abu Salamah F, Alatawneh I, Yakobson A. Melanoma Management: Exploring Staging, Prognosis, and Treatment Innovations. Int J Mol Sci 2024; 25:5794. [PMID: 38891988 PMCID: PMC11171767 DOI: 10.3390/ijms25115794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
Melanoma, a malignant neoplasm originating from melanocytes, stands as one of the most prevalent cancers globally, ranking fifth in terms of estimated new cases in recent years. Its aggressive nature and propensity for metastasis pose significant challenges in oncology. Recent advancements have led to a notable shift towards targeted therapies, driven by a deeper understanding of cutaneous tumor pathogenesis. Immunotherapy and tyrosine kinase inhibitors have emerged as promising strategies, demonstrating the potential to improve clinical outcomes across all disease stages, including neoadjuvant, adjuvant, and metastatic settings. Notably, there has been a groundbreaking development in the treatment of brain metastasis, historically associated with poor prognosis in oncology but showcasing impressive results in melanoma patients. This review article provides a comprehensive synthesis of the most recent knowledge on staging and prognostic factors while highlighting emerging therapeutic modalities, with a particular focus on neoadjuvant and adjuvant strategies, notably immunotherapy and targeted therapies, including the ongoing trials.
Collapse
Affiliation(s)
- Walid Shalata
- The Legacy Heritage Cancer Center and Larry Norton Institute, Soroka Medical Center, Beer Sheva 84105, Israel
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Zoe Gabrielle Attal
- Medical School for International Health, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Adam Solomon
- Medical School for International Health, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Sondos Shalata
- Nutrition Unit, Galilee Medical Center, Nahariya 22000, Israel
| | - Omar Abu Saleh
- Department of Dermatology and Venereology, The Emek Medical Centre, Afula 18341, Israel
| | - Lena Tourkey
- The Legacy Heritage Cancer Center and Larry Norton Institute, Soroka Medical Center, Beer Sheva 84105, Israel
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Fahed Abu Salamah
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
- Department of Dermatology, Soroka Medical Center, Beer Sheva 84105, Israel
| | - Ibrahim Alatawneh
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
- Department of Dermatology, Soroka Medical Center, Beer Sheva 84105, Israel
| | - Alexander Yakobson
- The Legacy Heritage Cancer Center and Larry Norton Institute, Soroka Medical Center, Beer Sheva 84105, Israel
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| |
Collapse
|
243
|
Janssen JC, van Dijk B, de Joode K, Aarts MJB, van den Berkmortel FWPJ, Blank CU, Boers-Sonderen MJ, van den Eertwegh AJM, de Groot JWB, Jalving M, de Jonge MJA, Joosse A, Kapiteijn E, Kamphuis-Huismans AM, Naipal KAT, Piersma D, Rikhof B, Westgeest HM, Vreugdenhil G, Oomen-de Hoop E, Mulder EEAP, van der Veldt AAM. Safe Stop IPI-NIVO trial: early discontinuation of nivolumab upon achieving a complete or partial response in patients with irresectable stage III or metastatic melanoma treated with first-line ipilimumab-nivolumab - study protocol. BMC Cancer 2024; 24:632. [PMID: 38783238 PMCID: PMC11112744 DOI: 10.1186/s12885-024-12336-0] [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: 01/30/2024] [Accepted: 05/03/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Patients with irresectable stage III or metastatic melanoma presenting with poor prognostic factors are usually treated with a combination of immune checkpoint inhibitors (ICIs), consisting of ipilimumab and nivolumab. This combination therapy is associated with severe immune related adverse events (irAEs) in about 60% of patients. In current clinical practice, patients are usually treated with ICIs for up to two years or until disease progression or the occurrence of unacceptable AEs. The incidence of irAEs gradually increases with duration of treatment. While durable tumour responses have been observed after early discontinuation of treatment, no consensus has been reached on optimal treatment duration. The objective of the Safe Stop IPI-NIVO trial is to evaluate whether early discontinuation of ICIs is safe in patients with irresectable stage III or metastatic melanoma who are treated with combination therapy. METHODS The Safe Stop IPI-NIVO trial is a nationwide, multicentre, prospective, single-arm, interventional study in the Netherlands. A total of 80 patients with irresectable stage III or metastatic melanoma who are treated with combination therapy of ipilimumab-nivolumab and have a complete or partial response (CR/PR) according to RECIST v1.1 will be included to early discontinue maintenance therapy with anti-PD-1. The primary endpoint is the rate of ongoing response at 12 months after start of ICI. Secondary endpoints include ongoing response at 24 months, disease control at different time points, melanoma specific and overall survival, the incidence of irAEs and health-related quality of life. DISCUSSION From a medical, healthcare and economic perspective, overtreatment should be prevented and shorter treatment duration of ICIs is preferred. If early discontinuation of ICIs is safe for patients who are treated with the combination of ipilimumab-nivolumab, the treatment duration of nivolumab could be shortened in patients with a favourable tumour response. TRIAL REGISTRATION ClinicalTrials.gov ID NCT05652673, registration date: 08-12-2022.
Collapse
Affiliation(s)
- J C Janssen
- Department of Medical Oncology and Radiology and Nuclear Medicine, Erasmus Medical Centre Cancer Institute, Dr. Molewaterplein 40, Rotterdam, 3015GD, The Netherlands
- Department of Surgical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - B van Dijk
- Department of Medical Oncology and Radiology and Nuclear Medicine, Erasmus Medical Centre Cancer Institute, Dr. Molewaterplein 40, Rotterdam, 3015GD, The Netherlands
| | - K de Joode
- Department of Medical Oncology and Radiology and Nuclear Medicine, Erasmus Medical Centre Cancer Institute, Dr. Molewaterplein 40, Rotterdam, 3015GD, The Netherlands
| | - M J B Aarts
- Department of Medical Oncology, Maastricht UMC+, Maastricht, The Netherlands
| | | | - C U Blank
- Department of Medical Oncology, NKI-AvL, Amsterdam, The Netherlands
- Department of Medical Oncology, Leiden UMC, Leiden, The Netherlands
| | | | - A J M van den Eertwegh
- Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - J W B de Groot
- Department of Medical Oncology, Isala Zwolle, Zwolle, The Netherlands
| | - M Jalving
- Department of Medical Oncology, UMC Groningen, Groningen, The Netherlands
| | - M J A de Jonge
- Department of Medical Oncology and Radiology and Nuclear Medicine, Erasmus Medical Centre Cancer Institute, Dr. Molewaterplein 40, Rotterdam, 3015GD, The Netherlands
| | - A Joosse
- Department of Medical Oncology and Radiology and Nuclear Medicine, Erasmus Medical Centre Cancer Institute, Dr. Molewaterplein 40, Rotterdam, 3015GD, The Netherlands
| | - E Kapiteijn
- Department of Medical Oncology, Leiden UMC, Leiden, The Netherlands
| | | | - K A T Naipal
- Department of Medical Oncology and Radiology and Nuclear Medicine, Erasmus Medical Centre Cancer Institute, Dr. Molewaterplein 40, Rotterdam, 3015GD, The Netherlands
| | - D Piersma
- Department of Medical Oncology, Medisch Spectrum Twente, Enschede, The Netherlands
| | - B Rikhof
- Department of Medical Oncology, Medisch Centrum Leeuwarden, Leeuwarden, The Netherlands
| | - H M Westgeest
- Department of Medical Oncology, Amphia Ziekenhuis, Breda, The Netherlands
| | - G Vreugdenhil
- Department of Medical Oncology, Maxima Medisch Centrum Veldhoven, Veldhoven, The Netherlands
| | - E Oomen-de Hoop
- Department of Medical Oncology and Radiology and Nuclear Medicine, Erasmus Medical Centre Cancer Institute, Dr. Molewaterplein 40, Rotterdam, 3015GD, The Netherlands
| | - E E A P Mulder
- Department of Medical Oncology and Radiology and Nuclear Medicine, Erasmus Medical Centre Cancer Institute, Dr. Molewaterplein 40, Rotterdam, 3015GD, The Netherlands
- Department of Surgical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Astrid A M van der Veldt
- Department of Medical Oncology and Radiology and Nuclear Medicine, Erasmus Medical Centre Cancer Institute, Dr. Molewaterplein 40, Rotterdam, 3015GD, The Netherlands.
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
244
|
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
|
245
|
Zhang X, Li J, Yang L, Zhu Y, Gao R, Zhang T, Chen X, Fu J, He G, Shi H, Peng S, Wu X. Targeted proteomics-determined multi-biomarker profiles developed classifier for prognosis and immunotherapy responses of advanced cervical cancer. Front Immunol 2024; 15:1391524. [PMID: 38835778 PMCID: PMC11148239 DOI: 10.3389/fimmu.2024.1391524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 04/30/2024] [Indexed: 06/06/2024] Open
Abstract
Background Cervical cancer (CC) poses a global health challenge, with a particularly poor prognosis in cases of recurrence, metastasis, or advanced stages. A single biomarker is inadequate to predict CC prognosis or identify CC patients likely to benefit from immunotherapy, presumably owing to tumor complexity and heterogeneity. Methods Using advanced Olink proteomics, we analyzed 92 oncology-related proteins in plasma from CC patients receiving immunotherapy, based upon the comparison of protein expression levels of pre-therapy with those of therapy-Cycle 6 in the partial response (PR) group and progressive disease (PD) group, respectively. Results 55 proteins were identified to exhibit differential expression trends across pre-therapy and post-therapy in both PR and PD groups. Enriched GO terms and KEGG pathways were associated with vital oncological and immunological processes. A logistic regression model, using 5 proteins (ITGB5, TGF-α, TLR3, WIF-1, and ERBB3) with highest AUC values, demonstrated good predictive performance for prognosis of CC patients undergoing immunotherapy and showed potential across different cancer types. The effectiveness of these proteins in prognosis prediction was further validated using TCGA-CESC datasets. A negative correlation and previously unidentified roles of WIF-1 in CC immunotherapy was also first determined. Conclusion Our findings reveal multi-biomarker profiles effectively predicting CC prognosis and identifying patients benefitting most from immunotherapy, especially for those with limited treatment options and traditionally poor prognosis, paving the way for personalized immunotherapeutic treatments and improved clinical strategies.
Collapse
Affiliation(s)
- Xu Zhang
- NHC Key Laboratory of Reproduction Regulation, Shanghai Engineering Research Center of Reproductive Health Drug and Devices, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Laboratory of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Jin Li
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Liuke Yang
- College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - Youwei Zhu
- Clinical Center of Bio-Therapy at Zhongshan Hospital & Institutes of Biomedical Sciences, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Rongrong Gao
- Clinical Center for Biotherapy at Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tiancheng Zhang
- NHC Key Laboratory of Reproduction Regulation, Shanghai Engineering Research Center of Reproductive Health Drug and Devices, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Laboratory of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Xuwen Chen
- Shanghai Kelin Clinical Bioinformatics Institute, Shanghai, China
| | - Jun Fu
- LC-Bio Technology Co., Ltd, Hangzhou, China
| | - Gaoyang He
- LC-Bio Technology Co., Ltd, Hangzhou, China
| | - Huijuan Shi
- NHC Key Laboratory of Reproduction Regulation, Shanghai Engineering Research Center of Reproductive Health Drug and Devices, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Laboratory of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Shenjie Peng
- Shanghai Medical College of Fudan University, Fudan University, Shanghai, China
| | - XiaoHua Wu
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
246
|
Ali DS, Gad HA, Hathout RM. Enhancing Effector Jurkat Cell Activity and Increasing Cytotoxicity against A549 Cells Using Nivolumab as an Anti-PD-1 Agent Loaded on Gelatin Nanoparticles. Gels 2024; 10:352. [PMID: 38920901 PMCID: PMC11202840 DOI: 10.3390/gels10060352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 05/11/2024] [Accepted: 05/17/2024] [Indexed: 06/27/2024] Open
Abstract
The current research investigated the use of gelatin nanoparticles (GNPs) for enhancing the cytotoxic effects of nivolumab, an immune checkpoint inhibitor. The unique feature of GNPs is their biocompatibility and functionalization potential, improving the delivery and the efficacy of immunotherapeutic drugs with fewer side effects compared to traditional treatments. This exploration of GNPs represents an innovative direction in the advancement of nanomedicine in oncology. Nivolumab-loaded GNPs were prepared and characterized. The optimum formulation had a particle size of 191.9 ± 0.67 nm, a polydispersity index of 0.027 ± 0.02, and drug entrapment of 54.67 ± 3.51%. A co-culture experiment involving A549 target cells and effector Jurkat cells treated with free nivolumab solution, and nivolumab-loaded GNPs, demonstrated that the latter had significant improvements in inhibition rate by scoring 87.88 ± 2.47% for drug-loaded GNPs against 60.53 ± 3.96% for the free nivolumab solution. The nivolumab-loaded GNPs had a lower IC50 value, of 0.41 ± 0.01 µM, compared to free nivolumab solution (1.22 ± 0.37 µM) at 72 h. The results indicate that administering nivolumab-loaded GNPs augmented the cytotoxicity against A549 cells by enhancing effector Jurkat cell activity compared to nivolumab solution treatment.
Collapse
Affiliation(s)
- Dalia S. Ali
- Department of Biotechnology, Central Administration of Biological, Innovative Products and Clinical Studies, Egyptian Drug Authority, Giza 11566, Egypt
| | - Heba A. Gad
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
- Pharmacy Program, Department of Pharmaceutical Sciences, Batterjee Medical College, Jeddah 21442, Saudi Arabia
| | - Rania M. Hathout
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
| |
Collapse
|
247
|
Sharma A, Alexander G, Chu JH, Markopoulos A, Maloul G, Ayub MT, Fidler MJ, Okwuosa TM. Immune Checkpoint Inhibitors and Cardiotoxicity: A Comparative Meta-Analysis of Observational Studies and Randomized Controlled Trials. J Am Heart Assoc 2024; 13:e032620. [PMID: 38761070 PMCID: PMC11179795 DOI: 10.1161/jaha.123.032620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 02/20/2024] [Indexed: 05/20/2024]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have uncommon associations with cardiotoxicity, yet these cardiotoxic effects are associated with high mortality. An accurate assessment of risk for cardiotoxicity is essential for clinical decision-making, but data from randomized controlled trials often differ from real-world observational studies. METHODS AND RESULTS A systematic search of PubMed, Embase, Cochrane Library, and Scopus was performed, including phase II and III randomized controlled trials (RCTs) and observational studies (OSs) reporting myocarditis or pericardial disease, myocardial infarction, or stroke with an immunotherapy. Odds ratios (ORs) were used to pool results between ICIs and other cancer therapy in RCTs and OSs. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses guideline was followed. In total, 54 RCTs (N=38 264) and 24 OSs (N=12 561 455) were included. In RCTs, ICI use resulted in higher risk of myocarditis (OR, 3.55 [95% CI, 2.10-5.98]), pericardial disease (OR, 2.73 [95% CI, 1.57-4.77]), and myocardial infarction (OR, 1.83 [95% CI, 1.03-3.25]), compared with non-ICI (placebo or chemotherapy). In OSs, ICI use was not associated with myocarditis, pericardial disease, or myocardial infarction compared with controls; however, combination ICIs demonstrated higher risk of myocarditis compared with single ICI use (OR, 3.07 [95% CI, 1.28-7.39]). Stroke risk was not increased with use of ICIs in RCTs. CONCLUSIONS We demonstrated increased risk of ICI myocarditis, pericardial disease, and myocardial infarction in RCTs but not OSs. Results of this study suggest there are differences between ICI cardiotoxicity risk, possibly suggesting differences in diagnoses and management, in clinical trials versus the OSs.
Collapse
Affiliation(s)
- Akash Sharma
- Department of Medicine University at Buffalo-Catholic Health System Buffalo NY
- Center for Global Health Research Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University Chennai India
| | - Grace Alexander
- Department of Internal Medicine University of Iowa Hospitals & Clinics Iowa City IA
| | - Jian H Chu
- Division of Cardiology, Department of Medicine University of Oklahoma Oklahoma City OK
| | | | | | - Muhammad Talha Ayub
- Heart and Vascular Institute, University of Pittsburgh Medical Center Pittsburgh PA
| | - Mary J Fidler
- Division of Hematology/Oncology/Stem cell transplant Rush University Medical Center Chicago IL
| | - Tochukwu M Okwuosa
- Division of Cardiology, Department of Internal Medicine Rush University Medical Center Chicago IL
| |
Collapse
|
248
|
Kadono T, Yamamoto S, Kato K. Development of perioperative immune checkpoint inhibitor therapy for locally advanced esophageal squamous cell carcinoma. Future Oncol 2024; 20:2097-2107. [PMID: 38861290 PMCID: PMC11497952 DOI: 10.1080/14796694.2024.2345043] [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: 05/24/2023] [Accepted: 04/16/2024] [Indexed: 06/12/2024] Open
Abstract
The standard preoperative treatment for resectable locally advanced esophageal squamous cell carcinoma (ESCC) is chemoradiotherapy in western countries (based on the CROSS trial) and triplet chemotherapy in Japan (based on the JCOG1109 trial). Postoperative nivolumab has recently been shown to improve disease-free survival in resectable locally advanced esophageal cancer after preoperative chemoradiotherapy in patients who had residual pathological disease, based on the CheckMate 577 trial. Furthermore, preoperative immune checkpoint inhibitor-containing treatments have also been developed. The JCOG1804E trial is presently evaluating the safety and efficacy of preoperative nivolumab-containing chemotherapy for resectable locally advanced ESCC. This review discusses the treatment of resectable locally advanced ESCC and future perspectives on perioperative immune checkpoint inhibitor-containing treatments.
Collapse
Affiliation(s)
- Toru Kadono
- Department of Head & Neck, Esophageal Medical Oncology, National Cancer Center Hospital, Tokyo, 104-0045, Japan
- Cancer Chemotherapy Center, Osaka Medical & Pharmaceutical University, Takatsuki, Osaka, 569-8686, Japan
| | - Shun Yamamoto
- Department of Head & Neck, Esophageal Medical Oncology, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Ken Kato
- Department of Head & Neck, Esophageal Medical Oncology, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| |
Collapse
|
249
|
Wang R, Chen Y, Xie Y, Ma X, Liu Y. Deciphering and overcoming Anti-PD-1 resistance in Melanoma: A comprehensive review of Mechanisms, biomarker Developments, and therapeutic strategies. Int Immunopharmacol 2024; 132:111989. [PMID: 38583243 DOI: 10.1016/j.intimp.2024.111989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 03/22/2024] [Accepted: 03/29/2024] [Indexed: 04/09/2024]
Abstract
Worldwide, tens of thousands of people die from melanoma each year, making it the most frequently fatal form of cutaneous cancer. Immunotherapeutic advancements, particularly with anti-PD-1 medications, have significantly enhanced treatment outcomes over recent decades. With the broad application of anti-PD-1 therapies, insights into the mechanisms of resistance have evolved. Despite the development of combination treatments and early predictive biomarkers, a comprehensive synthesis of these advancements is absent in the current literature. This review underscores the prevailing knowledge of anti-PD-1 resistance mechanisms and underscores the critical role of robust predictive biomarkers in stratifying patients for targeted combinations of anti-PD-1 and other conventional or innovative therapeutic approaches. Additionally, we offer insights that may shape future melanoma treatment strategies.
Collapse
Affiliation(s)
- Ruoqi Wang
- Shanghai Skin Disease Hospital, Shanghai Clinical College of Dermatology, Fifth Clinical Medical College, Anhui Medical University, Shanghai 200443, China
| | - Yanbin Chen
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai 200443, China
| | - Yongyi Xie
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai 200443, China
| | - Xin Ma
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai 200443, China; Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China.
| | - Yeqiang Liu
- Shanghai Skin Disease Hospital, Shanghai Clinical College of Dermatology, Fifth Clinical Medical College, Anhui Medical University, Shanghai 200443, China; Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai 200443, China.
| |
Collapse
|
250
|
Hou J, Xie S, Gao J, Jiang T, Zhu E, Yang X, Jin Z, Long H, Zhang A, Yang F, Wang L, Zha H, Jia Q, Zhu B, Wang X. NK cell transfer overcomes resistance to PD-(L)1 therapy in aged mice. Exp Hematol Oncol 2024; 13:48. [PMID: 38725070 PMCID: PMC11080179 DOI: 10.1186/s40164-024-00511-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 04/08/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Cancer is the leading cause of death among older adults. Although the integration of immunotherapy has revolutionized the therapeutic landscape of cancer, the complex interactions between age and immunotherapy efficacy remain incompletely defined. Here, we aimed to elucidate the relationship between aging and immunotherapy resistance. METHODS Flow cytometry was performed to evaluate the infiltration of immune cells in the tumor microenvironment (TME). In vivo T cell proliferation, cytotoxicity and migration assays were performed to evaluate the antitumor capacity of tumor antigen-specific CD8+ T cells in mice. Real-time quantitative PCR (qPCR) was used to investigate the expression of IFN-γ-associated gene and natural killer (NK)-associated chemokine. Adoptive NK cell transfer was adopted to evaluate the effects of NK cells from young mice in overcoming the immunotherapy resistance of aged mice. RESULTS We found that elderly patients with advanced non-small cell lung cancer (aNSCLC) aged ≥ 75 years exhibited poorer progression-free survival (PFS), overall survival (OS) and a lower clinical response rate after immunotherapy. Mechanistically, we showed that the infiltration of NK cells was significantly reduced in aged mice compared to younger mice. Furthermore, the aged NK cells could also suppress the activation of tumor antigen-specific CD8+ T cells by inhibiting the recruitment and activation of CD103+ dendritic cells (DCs). Adoptive transfer of NK cells from young mice to aged mice promoted TME remodeling, and reversed immunotherapy resistance. CONCLUSION Our findings revealed the decreased sensitivity of elderly patients to immunotherapy, as well as in aged mice. This may be attributed to the reduction of NK cells in aged mice, which inhibits CD103+ DCs recruitment and its CD86 expression and ultimately leads to immunotherapy resistance.
Collapse
Affiliation(s)
- Junlei Hou
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Shuanglong Xie
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
- Jinfeng Laboratory, Chongqing, 401329, China
| | - Jianbao Gao
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Tao Jiang
- Shanghai Pulmonary Hospital, Shanghai, 200082, China
| | - Enjian Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Xuezhi Yang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Zheng Jin
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Haixia Long
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Anmei Zhang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Fei Yang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Lujing Wang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Haoran Zha
- Department of Oncology, PLA Rocket Force Characteristic Medical Center, Beijing, 100088, China
| | - Qingzhu Jia
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Xinxin Wang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| |
Collapse
|