1
|
Idris OA, Westgate D, Saadaie Jahromi B, Shebrain A, Zhang T, Ashour HM. PD-L1 Inhibitor Cosibelimab for Cutaneous Squamous Cell Carcinoma: Comprehensive Evaluation of Efficacy, Mechanism, and Clinical Trial Insights. Biomedicines 2025; 13:889. [PMID: 40299523 PMCID: PMC12024788 DOI: 10.3390/biomedicines13040889] [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: 12/25/2024] [Revised: 03/18/2025] [Accepted: 03/19/2025] [Indexed: 04/30/2025] Open
Abstract
Cutaneous squamous cell carcinoma (CSCC) is one of the most common non-melanoma skin cancers, and particularly challenging to treat in advanced or metastatic stages. Traditional therapies, including chemotherapy and radiation, often result in limited efficacy and severe side effects. Cosibelimab, a fully human monoclonal antibody targeting PD-L1, has emerged as a promising immunotherapy for advanced CSCC. In this review, we evaluate the therapeutic potential of cosibelimab by analyzing its mechanism of action, clinical trial data, and its role compared to other PD-1/PD-L1 inhibitors, such as pembrolizumab and cemiplimab. We synthesized the available preclinical and clinical data on cosibelimab, focusing on published Phase I and II trial results involving 76 patients. Objective response rates (ORRs), progression-free survival (PFS), overall survival (OS), and safety profiles were compared between cosibelimab, pembrolizumab, and cemiplimab. Mechanistic insights into cosibelimab's dual action, including PD-L1 blockade and antibody-dependent cellular cytotoxicity (ADCC), were also explored. Phase II trials demonstrated an ORR of 47.5%, with a median PFS of 12.9 months in advanced CSCC patients. Cosibelimab demonstrated a favorable safety profile, with predominantly mild to moderate adverse events. Comparative analysis with pembrolizumab and cemiplimab showed similar efficacy, although long-term survival data for cosibelimab is still emerging. Given its efficacy and safety, cosibelimab holds promise not only as a monotherapy but also for future exploration in combination regimens and broader oncologic indications. Future trials are required to validate its long-term outcomes, including overall survival, and to explore its use in combination therapies and neoadjuvant/adjuvant settings.
Collapse
Affiliation(s)
- Omer A. Idris
- Department of Biological Sciences, Western Michigan University, Kalamazoo, MI 49008, USA; (O.A.I.); (A.S.)
- Malate Institute for Medical Research, Malate Inc., Grandville, MI 49468, USA
| | - Diana Westgate
- Homer Stryker MD School of Medicine, Western Michigan University, Kalamazoo, MI 49008, USA
- Forefront Dermatology, Kalamazoo, MI 49007, USA
| | - Bahar Saadaie Jahromi
- Department of Biological Sciences, Western Michigan University, Kalamazoo, MI 49008, USA; (O.A.I.); (A.S.)
| | - Abdulaziz Shebrain
- Department of Biological Sciences, Western Michigan University, Kalamazoo, MI 49008, USA; (O.A.I.); (A.S.)
| | - Tiantian Zhang
- Department of Hematology and Hematopoietic Stem Cell Transplantation, Toni Stephenson Lymphoma Center, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Hossam M. Ashour
- Department of Integrative Biology, College of Arts and Sciences, University of South Florida, St. Petersburg, FL 33701, USA
| |
Collapse
|
2
|
Huang R, Shen ZY, Huang D, Zhao SH, Dan LX, Wu P, Tang QZ, Ma ZG. Microbiota-indole-3-propionic acid-heart axis mediates the protection of leflunomide against αPD1-induced cardiotoxicity in mice. Nat Commun 2025; 16:2651. [PMID: 40108157 PMCID: PMC11923180 DOI: 10.1038/s41467-025-58107-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 03/12/2025] [Indexed: 03/22/2025] Open
Abstract
Anti-programmed death 1 (αPD1) immune checkpoint blockade is used in combination for cancer treatment but associated with cardiovascular toxicity. Leflunomide (Lef) can suppress the growth of several tumor and mitigate cardiac remodeling in mice. However, the role of Lef in αPD1-induced cardiotoxicity remains unclear. Here, we report that Lef treatment inhibits αPD1-related cardiotoxicity without compromising the efficacy of αPD1-mediated immunotherapy. Lef changes community structure of gut microbiota in αPD1-treated melanoma-bearing mice. Moreover, mice receiving microbiota transplants from Lef+αPD1-treated melanoma-bearing mice have better cardiac function compared to mice receiving transplants from αPD1-treated mice. Mechanistically, we analyze metabolomics and identify indole-3-propionic acid (IPA), which protects cardiac dysfunction in αPD1-treated mice. IPA can directly bind to the aryl hydrocarbon receptor and promote phosphoinositide 3-kinase expression, thus curtailing the cardiomyocyte response to immune injury. Our findings reveal that Lef mitigates αPD1-induced cardiac toxicity in melanoma-bearing mice through modulation of the microbiota-IPA-heart axis.
Collapse
Affiliation(s)
- Rong Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Zhuo-Yu Shen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Dan Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Shu-Hong Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Ling-Xuan Dan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Pan Wu
- Department of Adult Internal Medicine, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Zhen-Guo Ma
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China.
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China.
| |
Collapse
|
3
|
Taghizadeh H, Fajkovic H. Immunotherapy in the Management of Penile Cancer-A Systematic Review. Cancers (Basel) 2025; 17:883. [PMID: 40075730 PMCID: PMC11898862 DOI: 10.3390/cancers17050883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/22/2025] [Accepted: 03/02/2025] [Indexed: 03/14/2025] Open
Abstract
Penile cancer, though a rare malignancy, presents a significant challenge in the domain of male genitourinary oncology, particularly due to its limited treatment options and due to the profound physical and psychological impact on patients [...].
Collapse
Affiliation(s)
- Hossein Taghizadeh
- Division of Oncology, Department of Internal Medicine I, University Hospital St. Pölten, 3100 St. Pölten, Austria;
- Divison of Oncology, Karl Landsteiner University of Health Sciences, 3500 Krems, Austria
- Karl Landsteiner Institute for Oncology and Nephrology, Dunant-Platz 1, 3100 St. Pölten, Austria
| | - Harun Fajkovic
- Divison of Oncology, Karl Landsteiner University of Health Sciences, 3500 Krems, Austria
- Department of Urology and Andrology, University Hospital St. Pölten, 3100 St. Pölten, Austria
- Karl Landsteiner Institute for Urological Research and Training, 3100 St. Pölten, Austria
- Comprehensive Cancer Center, Department of Urology, Medical University of Vienna, Vienna General Hospital, 1090 Vienna, Austria
| |
Collapse
|
4
|
Zhang X, Lan R, Liu Y, Pillarisetty VG, Li D, Zhao CL, Sarkar SA, Liu W, Hanna I, Gupta M, Hajdu C, Melamed J, Shusterman M, Widmer J, Allendorf J, Liu YZ. Complement activation in tumor microenvironment after neoadjuvant therapy and its impact on pancreatic cancer outcomes. NPJ Precis Oncol 2025; 9:58. [PMID: 40032924 PMCID: PMC11876354 DOI: 10.1038/s41698-025-00848-2] [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: 03/14/2024] [Accepted: 02/24/2025] [Indexed: 03/05/2025] Open
Abstract
Neoadjuvant therapy (NAT) is increasingly being used for pancreatic ductal adenocarcinoma (PDAC). This study investigates how NAT differentially impacts PDAC's carcinoma cells and the tumor microenvironment (TME). Spatial transcriptomics was used to compare gene expression profiles in carcinoma cells and the TME of 23 NAT-treated versus 13 NAT-naïve PDACs. Findings were validated by single-nucleus RNA sequencing (snRNA-seq) analysis. NAT induces apoptosis and inhibits proliferation of carcinoma cells and coordinately upregulates multiple complement genes (C1R, C1S, C3, C4B and C7) within the TME. Higher TME complement expression following NAT is associated with increased immunomodulatory and neurotrophic cancer-associated fibroblasts (CAFs); more CD4+ T cells; reduced immune exhaustion gene expression, and improved overall survival. snRNA-seq analysis demonstrates C3 complement is mainly upregulated in CAFs. These findings suggest that local complement dynamics could serve as a novel biomarker for prognosis, evaluating treatment response, and guiding therapeutic strategies in NAT-treated PDAC patients.
Collapse
Affiliation(s)
- Xiaofei Zhang
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Pathology and Laboratory Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY, USA.
| | - Ruoxin Lan
- Department of Biostatistics and Data Science, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Yongjun Liu
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - Venu G Pillarisetty
- Department of Surgery, University of Washington School of Medicine, Seattle, WA, USA
| | - Danting Li
- Department of Biostatistics and Data Science, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Chaohui L Zhao
- Department of Pathology and Laboratory Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY, USA
| | - Suparna A Sarkar
- Department of Pathology and Laboratory Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Weiguo Liu
- Department of Pathology and Laboratory Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY, USA
| | - Iman Hanna
- Department of Pathology and Laboratory Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY, USA
| | - Mala Gupta
- Department of Pathology and Laboratory Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY, USA
| | - Cristina Hajdu
- Department of Pathology and Laboratory Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Jonathan Melamed
- Department of Pathology and Laboratory Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY, USA
| | - Michael Shusterman
- Department of Oncology, New York University Grossman Long Island School of Medicine, Mineola, NY, USA
| | - Jessica Widmer
- Department of Gastroenterology, New York University Grossman Long Island School of Medicine, Mineola, NY, USA
| | - John Allendorf
- Department of Surgery, New York University Grossman Long Island School of Medicine, Mineola, NY, USA
| | - Yao-Zhong Liu
- Department of Biostatistics and Data Science, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA.
| |
Collapse
|
5
|
Uher O, Hadrava Vanova K, Labitt R, Petrlakova K, Ye J, Wang H, Masarik M, Jakubek M, Zenka J, Zhuang Z, Pacak K. Neoadjuvant intratumoral MBT(A) immunotherapy prevents distant metastases and recurrence in murine models. Cancer Lett 2025; 612:217464. [PMID: 39809356 DOI: 10.1016/j.canlet.2025.217464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 12/15/2024] [Accepted: 01/11/2025] [Indexed: 01/16/2025]
Abstract
Neoadjuvant immunotherapy represents a pioneering approach in the preoperative treatment of cancer, providing new strategies for tumor reduction and improved patient outcomes by modulating the immune response. This study investigated neoadjuvant immunotherapy using intratumoral administration of mannan-BAM, Toll-like receptor ligands, and anti-CD40 antibody (MBTA therapy) followed by surgery in murine models of MTT pheochromocytoma, B16-F10 melanoma, and 4T1 and E0771.lmb mammary carcinomas. In the MTT pheochromocytoma model, it was found that neoadjuvant MBTA therapy followed by surgery could prevent the development of distant metastases in 100% of treated animals, compared to a 60% mortality rate in the control group due to metastatic disease after surgery. These outcomes were achieved even in tumors three times larger than those in the control group. In the aggressive 4T1 model, neoadjuvant MBTA therapy resulted in slower tumor progression and a significant prolongation of survival. In the B16-F10 and E0771.lmb models, neoadjuvant MBTA therapy also protected animals from metastases development and tumor recurrence upon rechallenge with tumor cells after surgery. Transcriptomic analysis revealed enhanced effector immune cell infiltration, cytotoxicity, and antigen presentation in retransplanted tumors from MBTA-treated mice, indicating robust immune memory. Notably, the exclusion of the anti-CD40 antibody from the neoadjuvant MBTA therapy (MBT therapy) yielded comparable outcomes in protection against metastases development. These findings advocate for further investigation of intratumoral neoadjuvant MBTA therapy for immunologically "cold" tumors, including those at high risk of metastases or recurrence.
Collapse
MESH Headings
- Animals
- Female
- Neoadjuvant Therapy/methods
- Mice
- Immunotherapy/methods
- Mice, Inbred C57BL
- Neoplasm Recurrence, Local/prevention & control
- Neoplasm Recurrence, Local/immunology
- Cell Line, Tumor
- Melanoma, Experimental/immunology
- Melanoma, Experimental/therapy
- Melanoma, Experimental/pathology
- Melanoma, Experimental/secondary
- CD40 Antigens/immunology
- CD40 Antigens/antagonists & inhibitors
- Mice, Inbred BALB C
- Disease Models, Animal
- Neoplasm Metastasis
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/therapy
Collapse
Affiliation(s)
- Ondrej Uher
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA
| | - Katerina Hadrava Vanova
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA
| | - Rachael Labitt
- Research Animal Management Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA
| | - Katerina Petrlakova
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA; Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Juan Ye
- Neuro-Oncology Branch, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Herui Wang
- Neuro-Oncology Branch, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Michal Masarik
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic; Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic; BIOCEV (Biotechnology and Biomedicine Center in Vestec), First Faculty of Medicine, Charles University, Vestec, Czech Republic
| | - Milan Jakubek
- BIOCEV (Biotechnology and Biomedicine Center in Vestec), First Faculty of Medicine, Charles University, Vestec, Czech Republic; Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Jan Zenka
- Department of Medical Biology, Faculty of Science, University of South Bohemia, Ceske Budejovice, Czech Republic
| | - Zhengping Zhuang
- Neuro-Oncology Branch, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA; AKESO, Prague 5, Czech Republic.
| |
Collapse
|
6
|
Xu P, Yu H, Bian H, Jia D, Li W, Dong H, Peng X. Bibliometric insight into neoadjuvant immunotherapy in non-small cell lung cancer: trends, collaborations, and future avenues. Front Immunol 2025; 16:1533651. [PMID: 39995679 PMCID: PMC11847801 DOI: 10.3389/fimmu.2025.1533651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 01/21/2025] [Indexed: 02/26/2025] Open
Abstract
Background Neoadjuvant immunotherapy (NIT) is a rapidly emerging paradigm for advanced resectable non-small cell lung cancer (NSCLC). However, there is no bibliometric analysis in this research field. Objective To analyze the hotspots and trends in the research of NIT for NSCLC and provide a reference for the study of NIT for lung cancer in China. Methods Retrieve literature related to NIT for NSCLC from Web of Science, PubMed, and Scopus databases up to September 10, 2024. Use CiteSpace and VOSviewer software visualization software to analyze the keywords of country, author, institution, and literature. Results There were 1575 references, and the overall annual publication volume showed an upward trend; Forde and Patrick M have published the most articles in the literature. The research hotspots mainly focus on chemotherapy, NIT for NSCLC, immunotherapy, neoadjuvant chemotherapy, pathological reactions, etc. Conclusions This is the first bibliometric study comprehensively summarizing NIT's research trends and development in NSCLC. Our bibliometric assessment provides a panoramic view of the research milieu surrounding NIT for NSCLC, encapsulating the present state, evolving trends, and potential future directions, particularly emphasizing the promise of immunochemotherapy.
Collapse
Affiliation(s)
- Pengliang Xu
- Department of Thoracic Surgery, The First People’s Hospital of Huzhou, Huzhou, China
| | - Huanming Yu
- Department of Thoracic Surgery, The First People’s Hospital of Huzhou, Huzhou, China
| | - Hupo Bian
- Department of Radiology, The First People’s Hospital of Huzhou, Huzhou, China
| | - Dan Jia
- Department of Respiratory Medicine, The First People’s Hospital of Huzhou, Huzhou, China
| | - Wenhui Li
- Department of Thoracic Surgery, The First People’s Hospital of Huzhou, Huzhou, China
| | - Hongfeng Dong
- Department of Radiology, The First People’s Hospital of Huzhou, Huzhou, China
| | - Xiuhua Peng
- Department of Radiology, The First People’s Hospital of Huzhou, Huzhou, China
| |
Collapse
|
7
|
Sun Y, Yang G, Sun R, Cao F. Neoadjuvant immunotherapy plus chemotherapy for squamous cell carcinoma of the paranasal sinus: a case report. Front Oncol 2024; 14:1462993. [PMID: 39777334 PMCID: PMC11703731 DOI: 10.3389/fonc.2024.1462993] [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: 07/11/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) such as pembrolizumab and nivolumab are recommended as first-line therapies for recurrent and metastatic head and neck squamous cell carcinoma (HNSCC). However, their efficacy in neoadjuvant therapy remains uncertain. Case presentation We report the case of a 68-year-old male diagnosed with HNSCC who received neoadjuvant nivolumab (anti-PD-1 inhibitor) plus nab-paclitaxel and carboplatin. Biomarkerswere assessed by immunohistochemistry, and apoptosis-related molecules were analyzed via Western blotting. The patient achieved significant tumor regression and major pathological response (MPR) without severe adverse events. Post-treatment analyses revealed PD-L1 expression increased from 30% to 50% in tumor cells, CD8+ lymphocyte infiltration significantly improved, and Ki-67 expression was markedly reduced. Conclusions This case highlights the potential of combining ICIs with chemotherapy in neoadjuvant settings for HNSCC, providing mechanistic insights and clinical evidence for this emerging approach. Further studies are needed to establish the optimal neoadjuvant treatment regimen and identify patient populations most likely to benefit.
Collapse
Affiliation(s)
- Ying Sun
- Department of Medical Oncology, Qilu Hospital of Shandong University (Qingdao), Qingdao, Shandong, China
| | - Guanghui Yang
- Department of Medical Oncology, Qilu Hospital of Shandong University (Qingdao), Qingdao, Shandong, China
| | - Ruijie Sun
- Department of Otolaryngology, Qilu Hospital of Shandong University (Qingdao), Qingdao, Shandong, China
| | - Fangli Cao
- Department of Medical Oncology, Qilu Hospital of Shandong University (Qingdao), Qingdao, Shandong, China
| |
Collapse
|
8
|
Cuiffo B, Maxwell M, Yan D, Guemiri R, Boone A, Bellet D, Rivest B, Cardia J, Robert C, Fricker SP. Self-delivering RNAi immunotherapeutic PH-762 silences PD-1 to generate local and abscopal antitumor efficacy. Front Immunol 2024; 15:1501679. [PMID: 39697325 PMCID: PMC11652358 DOI: 10.3389/fimmu.2024.1501679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 11/08/2024] [Indexed: 12/20/2024] Open
Abstract
Objective Immunotherapeutic inhibition of PD-1 by systemically administered monoclonal antibodies is widely used in cancer treatment, but it may cause severe immune-related adverse events (irSAEs). Neoadjuvant PD-1 inhibition before surgery has shown promise in reducing recurrence by stimulating durable antitumor immunity. Local intratumoral (IT) immunotherapy is a potential strategy to minimize irSAEs, but antibodies have limited tumor penetration, making them less suitable for this approach. Therapeutic self-delivering RNAi (INTASYL) is an emerging modality well-suited for neoadjuvant immunotherapy. This study presents preclinical proof-of-concept for PH-762, an INTASYL designed to silence PD-1, currently in clinical development for advanced cutaneous malignancies (ClinicalTrials.gov#NCT06014086). Methods and analysis PH-762 pharmacology was characterized in vitro, and in vivo antitumor efficacy was evaluated using a murine analogue (mPH-762) in syngeneic tumor models with varying PD-1 responsiveness. Bilateral Hepa1-6 models assessed abscopal effects of local treatment. Ex vivo analyses explored mechanisms of direct and abscopal efficacy. Results PH-762 was rapidly internalized by human T cells, silencing PD-1 mRNA and decreasing PD-1 surface protein, enhancing TCR-stimulated IFN-γ and CXCL10 secretion. In vivo, IT mPH-762 provided robust antitumor efficacy, local and lymphatic biodistribution, and was well tolerated. Ex vivo analyses revealed that IT mPH-762 depleted PD-1 protein, promoted leukocyte and T cell infiltration, and correlated with tumor control. IT mPH-762 also demonstrated efficacy against untreated distal tumors (abscopal effect) by priming systemic antitumor immunity. Conclusion These data support PH-762 as a promising candidate for neoadjuvant immunotherapy in clinical studies.
Collapse
Affiliation(s)
| | | | - Dingxue Yan
- Phio Pharmaceuticals, Marlborough, MA, United States
| | - Ramdane Guemiri
- Dermatology Unit, Gustave Roussy Cancer Center, Villejuif, France
| | - Andrew Boone
- Phio Pharmaceuticals, Marlborough, MA, United States
| | - Deborah Bellet
- Dermatology Unit, Gustave Roussy Cancer Center, Villejuif, France
| | | | - James Cardia
- Phio Pharmaceuticals, Marlborough, MA, United States
| | - Caroline Robert
- Dermatology Unit, Gustave Roussy Cancer Center, Villejuif, France
| | | |
Collapse
|
9
|
Lucas S, Thomas SN. Therapeutic Immunomodulation of Tumor-Lymphatic Crosstalk via Intratumoral Immunotherapy. Mol Pharm 2024; 21:5929-5943. [PMID: 39478434 PMCID: PMC11615947 DOI: 10.1021/acs.molpharmaceut.4c00692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 12/06/2024]
Abstract
Intra- and peritumoral lymphatics and tumor-draining lymph nodes play major roles in mediating the adaptive immune response to cancer immunotherapy. Despite this, current paradigms of clinical cancer management seldom seek to therapeutically modulate tumor-lymphatic immune crosstalk. This review explores recent developments that set the stage for how this regulatory axis can be therapeutically manipulated, with a particular emphasis on tumor-localized immunomodulation. Building on this idea, the nature of tumor-lymphatic immune crosstalk and relevant immunotherapeutic targets and pathways are reviewed, with a focus on their translational potential. Engineered drug delivery systems that enhance intratumoral immunotherapy by improving drug delivery to both the tumor and lymph nodes are also highlighted.
Collapse
Affiliation(s)
- Samuel
N. Lucas
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
of America
| | - Susan N. Thomas
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
of America
- George
W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States of America
- Parker
H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, United States of America
- Winship
Cancer Institute, Emory University, Atlanta, Georgia 30322, United States
of America
| |
Collapse
|
10
|
Palollathil A, Babu S, Abhinand CS, Mathew RT, Vijayakumar M, Prasad TSK. Proteomic profiling of oral squamous cell carcinoma tissues reveals altered immune-related proteins: implications for personalized therapy. Expert Rev Proteomics 2024; 21:483-495. [PMID: 39523852 DOI: 10.1080/14789450.2024.2428332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVES Oral squamous cell carcinoma poses a substantial global health challenge marked by rising mortality rate. Recently, immunotherapy has shown promising results in cancer management by enhancing immune response. Thus, identifying additional immune-related markers is critical for advancing immunotherapy treatments. METHODS Data-independent acquisition (DIA) mass spectrometry approach was used to explore differentially expressed immune-related proteins in oral cancer tissues compared to adjacent non-cancerous tissues. Functional significance was identified through Gene Ontology, pathway, and network analysis. Gene expression of identified proteins was validated using transcriptomic data. RESULTS DIA analysis identified 29,459 precursors corresponding to 3429 proteins. Among these, 1060 proteins were differentially expressed, with 166 being immune-related. Differentially regulated proteins were involved in innate immune response, mitochondrial ATP synthesis, and neutrophil degranulation. Pathway analysis of immune-related proteins showed perturbation in anti-tumor immunity-related pathways such as interferon signaling, TCR signaling, PD-1 signaling, and antigen processing and presentation. Significance of these pathways was further reinforced by the strong interactions identified in the protein-protein interaction network analysis. Additionally, gene expression analysis showed similar mRNA expression patterns for key proteins involved in altered pathways, including ISG15, IFIT1/3, HLA-A/C and OAS2/3. CONCLUSIONS Further validation of these proteins could establish them as potential targets for personalized therapy.
Collapse
Affiliation(s)
- Akhina Palollathil
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| | - Sreeranjini Babu
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| | - Chandran S Abhinand
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| | - Rohan Thomas Mathew
- Department of Surgical Oncology, Yenepoya Medical College Hospital, Yenepoya (Deemed to be University), Mangalore, India
| | - Manavalan Vijayakumar
- Department of Surgical Oncology, Yenepoya Medical College Hospital, Yenepoya (Deemed to be University), Mangalore, India
| | | |
Collapse
|
11
|
Guo W, Qiao T, Li H, Zhao Y, Qin J, Zhang C, Shi C. Peripheral CD8 +PD-1 + T cells as novel biomarker for neoadjuvant chemoimmunotherapy in humanized mice of non-small cell lung cancer. Cancer Lett 2024; 597:217073. [PMID: 38906523 DOI: 10.1016/j.canlet.2024.217073] [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: 02/14/2024] [Revised: 06/11/2024] [Accepted: 06/17/2024] [Indexed: 06/23/2024]
Abstract
Neoadjuvant immunotherapy has shown promising clinical activity in the treatment of early non-small cell lung cancer (NSCLC); however, further clarification of the specific mechanism and identification of biomarkers are imperative prior to implementing it as a daily practice. The study investigated the reprogramming of T cells in both tumor and peripheral blood following neoadjuvant chemoimmunotherapy in a preclinical NSCLC mouse model engrafted with a human immune system. Samples were also collected from 21 NSCLC patients (Stage IA-IIIB) who received neoadjuvant chemoimmunotherapy, and the dynamics of potential biomarkers within these samples were measured and further subjected to correlation analysis with prognosis. Further, we initially investigated the sources of the potential biomarkers. We observed in the humanized mouse model, neoadjuvant chemoimmunotherapy could prevent postoperative recurrence and metastasis by increasing the frequency and cytotoxicity of CD8+ T cells in both peripheral blood (p < 0.001) and tumor immune microenvironment (TIME) (p < 0.001). The kinetics of peripheral CD8+PD-1+ T cells reflected the changes in the TIME and pathological responses, ultimately predicting survival outcome of mice. In the clinical cohort, patients exhibiting an increase in these T cells post-treatment had a higher rate of complete or major pathological response (p < 0.05) and increased immune infiltration (p = 0.0012, r = 0.792). We identified these T cells originating from tumor draining lymph nodes and subsequently entering the TIME. In conclusion, the kinetics of peripheral CD8+PD-1+ T cells can serve as a predictor for changes in TIME and optimal timing for surgery, ultimately reflecting the outcomes of neoadjuvant chemoimmunotherapy in both preclinical and clinical setting.
Collapse
Affiliation(s)
- Wenwen Guo
- Division of Cancer Biology, Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Clinical Research Center, Xianyang Central Hospital, Xianyang, Shaanxi, 712099, China
| | - Tianyun Qiao
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Hui Li
- Division of Cancer Biology, Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Yong Zhao
- Division of Cancer Biology, Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Jing Qin
- Division of Cancer Biology, Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Caiqin Zhang
- Division of Cancer Biology, Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| | - Changhong Shi
- Division of Cancer Biology, Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
12
|
Uher O, Hadrava Vanova K, Taïeb D, Calsina B, Robledo M, Clifton-Bligh R, Pacak K. The Immune Landscape of Pheochromocytoma and Paraganglioma: Current Advances and Perspectives. Endocr Rev 2024; 45:521-552. [PMID: 38377172 PMCID: PMC11244254 DOI: 10.1210/endrev/bnae005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/19/2023] [Accepted: 02/02/2024] [Indexed: 02/22/2024]
Abstract
Pheochromocytomas and paragangliomas (PPGLs) are rare neuroendocrine tumors derived from neural crest cells from adrenal medullary chromaffin tissues and extra-adrenal paraganglia, respectively. Although the current treatment for PPGLs is surgery, optimal treatment options for advanced and metastatic cases have been limited. Hence, understanding the role of the immune system in PPGL tumorigenesis can provide essential knowledge for the development of better therapeutic and tumor management strategies, especially for those with advanced and metastatic PPGLs. The first part of this review outlines the fundamental principles of the immune system and tumor microenvironment, and their role in cancer immunoediting, particularly emphasizing PPGLs. We focus on how the unique pathophysiology of PPGLs, such as their high molecular, biochemical, and imaging heterogeneity and production of several oncometabolites, creates a tumor-specific microenvironment and immunologically "cold" tumors. Thereafter, we discuss recently published studies related to the reclustering of PPGLs based on their immune signature. The second part of this review discusses future perspectives in PPGL management, including immunodiagnostic and promising immunotherapeutic approaches for converting "cold" tumors into immunologically active or "hot" tumors known for their better immunotherapy response and patient outcomes. Special emphasis is placed on potent immune-related imaging strategies and immune signatures that could be used for the reclassification, prognostication, and management of these tumors to improve patient care and prognosis. Furthermore, we introduce currently available immunotherapies and their possible combinations with other available therapies as an emerging treatment for PPGLs that targets hostile tumor environments.
Collapse
Affiliation(s)
- Ondrej Uher
- Section of Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-1109, USA
| | - Katerina Hadrava Vanova
- Section of Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-1109, USA
| | - David Taïeb
- Department of Nuclear Medicine, CHU de La Timone, Marseille 13005, France
| | - Bruna Calsina
- Hereditary Endocrine Cancer Group, Human Cancer Genetics Program, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain
- Familiar Cancer Clinical Unit, Human Cancer Genetics Program, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain
| | - Mercedes Robledo
- Hereditary Endocrine Cancer Group, Human Cancer Genetics Program, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Institute of Health Carlos III (ISCIII), Madrid 28029, Spain
| | - Roderick Clifton-Bligh
- Department of Endocrinology, Royal North Shore Hospital, Sydney 2065, NSW, Australia
- Cancer Genetics Laboratory, Kolling Institute, University of Sydney, Sydney 2065, NSW, Australia
| | - Karel Pacak
- Section of Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-1109, USA
| |
Collapse
|
13
|
Czarnecka AM, Ostaszewski K, Błoński P, Szumera-Ciećkiewicz A, Kozak K, Placzke J, Borkowska A, Terlecka A, Rogala P, Świtaj T, Sałamacha M, Mitręga-Korab B, Krotewicz M, Dudzisz-Śledź M, Rutkowski P. Preoperative-postoperative immunotherapy as treatment of borderline resectable and oligoprogressive stage III B-D and IV melanoma. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024; 50:108382. [PMID: 38763112 DOI: 10.1016/j.ejso.2024.108382] [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: 11/19/2023] [Revised: 04/11/2024] [Accepted: 04/30/2024] [Indexed: 05/21/2024]
Abstract
INTRODUCTION Perioperative therapy has gained significant importance in patients with advanced melanoma. Currently, there is little data on the routine use of preoperative immunotherapy in metastatic melanoma outside clinical trials. This study aimed to evaluate the effectiveness of preoperative treatment in patients with borderline resectable stage III or IV melanoma as well as in oligoprogressing stage IV cases; the secondary aim is to describe the safety of surgery after immunotherapy. MATERIALS AND METHODS Since 1/Jan/2016 seventeen patients were treated with curative intent neoadjuvant immunotherapy, surgery, and adjuvant immunotherapy, while nineteen patients were operated due to oligoprogression while treted with immunotherapy. Survival was analyzed using the Kaplan-Meier method and association between variables was tested using the chi-squared test. RESULTS R0 resection was achieved in 76.5 % of cases after neoadjuvant immunotherapy. 24 % of patients achieved objective RECIST response and 35 % complete or major pathological response. At the median follow-up time of 51.4 months, 64.7 % of patients were free of PD after perioperative treatment, while 3-year RFS and OS rates were 68 % and 80.9 %, respectively. R0 resection was achieved in 73.7 % of oligo-progressing nodules. The median time to PD on immunotherapy after the first oligoprogression was 10.3 months. Immunotherapy did not result in any unexpected surgical complications. No patient died during preoperative treatment due to immunotherapy toxicity or disease progression. CONCLUSIONS We confirmed treatment safety and long-term disease control after perioperative immunotherapy. Patients with borderline resectable melanoma should be referred to reference centers using neoadjuvant immunotherapy.
Collapse
Affiliation(s)
- Anna M Czarnecka
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland.
| | - Krzysztof Ostaszewski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Piotr Błoński
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland; Faculty of Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Anna Szumera-Ciećkiewicz
- Department of Pathology and Laboratory Diagnostics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Katarzyna Kozak
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Joanna Placzke
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Aneta Borkowska
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland; Department of Radiology I, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Anna Terlecka
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland; Faculty of Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Paweł Rogala
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Tomasz Świtaj
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Maciej Sałamacha
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Beata Mitręga-Korab
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Maria Krotewicz
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Monika Dudzisz-Śledź
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| |
Collapse
|
14
|
Zhang X, Lan R, Liu Y, Pillarisetty VG, Li D, Zhao CL, Sarkar SA, Liu W, Hanna I, Gupta M, Hajdu C, Melamed J, Shusterman M, Widmer J, Allendorf J, Liu YZ. Enhanced Complement Expression in the Tumor Microenvironment Following Neoadjuvant Therapy: Implications for Immunomodulation and Survival in Pancreatic Ductal Adenocarcinoma. RESEARCH SQUARE 2024:rs.3.rs-4104258. [PMID: 38798691 PMCID: PMC11118688 DOI: 10.21203/rs.3.rs-4104258/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Background Neoadjuvant therapy (NAT) is increasingly being used for pancreatic ductal adenocarcinoma (PDAC) treatment. However, its specific effects on carcinoma cells and the tumor microenvironment (TME) are not fully understood. This study aims to investigate how NAT differentially impacts PDAC's carcinoma cells and TME. Methods Spatial transcriptomics was used to compare gene expression profiles in carcinoma cells and the TME between 23 NAT-treated and 13 NAT-naïve PDAC patients, correlating with their clinicopathologic features. Analysis of an online single-nucleus RNA sequencing (snRNA-seq) dataset was performed for validation of the specific cell types responsible for NAT-induced gene expression alterations. Results NAT not only induces apoptosis and inhibits proliferation in carcinoma cells but also significantly remodels the TME. Notably, NAT induces a coordinated upregulation of multiple key complement genes (C3, C1S, C1R, C4B and C7) in the TME, making the complement pathway one of the most significantly affected pathways by NAT. Patients with higher TME complement expression following NAT exhibit improved overall survival. These patients also exhibit increased immunomodulatory and neurotrophic cancer-associated fibroblasts (CAFs); more CD4+ T cells, monocytes, and mast cells; and reduced immune exhaustion gene expression. snRNA-seq analysis demonstrates C3 complement was specifically upregulated in CAFs but not in other stroma cell types. Conclusions NAT can enhance complement production and signaling within the TME, which is associated with reduced immunosuppression in PDAC. These findings suggest that local complement dynamics could serve as a novel biomarker for prognosis, evaluating treatment response and resistance, and guiding therapeutic strategies in NAT-treated PDAC patients.
Collapse
Affiliation(s)
- Xiaofei Zhang
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI
- Department of Pathology and Laboratory Medicine, New York University Grossman Long Island School of Medicine, Long Island, NY
| | - Ruoxin Lan
- Department of Biostatistics and Data Science, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA
| | - Yongjun Liu
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA
| | - Venu G Pillarisetty
- Department of Surgery, University of Washington School of Medicine, Seattle, WA
| | - Danting Li
- Department of Biostatistics and Data Science, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA
| | - Chaohui L Zhao
- Department of Pathology and Laboratory Medicine, New York University Grossman Long Island School of Medicine, Long Island, NY
| | - Suparna A. Sarkar
- Department of Pathology and Laboratory Medicine, New York University Grossman School of Medicine, New York, NY
| | - Weiguo Liu
- Department of Pathology and Laboratory Medicine, New York University Grossman Long Island School of Medicine, Long Island, NY
| | - Iman Hanna
- Department of Pathology and Laboratory Medicine, New York University Grossman Long Island School of Medicine, Long Island, NY
| | - Mala Gupta
- Department of Pathology and Laboratory Medicine, New York University Grossman Long Island School of Medicine, Long Island, NY
| | - Cristina Hajdu
- Department of Pathology and Laboratory Medicine, New York University Grossman School of Medicine, New York, NY
| | - Jonathan Melamed
- Department of Pathology and Laboratory Medicine, New York University Grossman Long Island School of Medicine, Long Island, NY
| | - Michael Shusterman
- Department of Oncology, New York University Grossman Long Island School of Medicine, Long Island, NY
| | - Jessica Widmer
- Department of Gastroenterology, New York University Grossman Long Island School of Medicine, Long Island, NY
| | - John Allendorf
- Department of Surgery, New York University Grossman Long Island School of Medicine, Long Island, NY
| | - Yao-Zhong Liu
- Department of Biostatistics and Data Science, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA
| |
Collapse
|
15
|
Zheng J, He Z, Shen L, Chen X, Chen P, Zhang B, Qin H, Xiong Z, Zhang S. Microwave-Responsive Edge-Oxidized Graphene for Imaging-Guided Neoadjuvant Thermal Immunotherapy via Promoting Immunogenic Cell Death and Redressing Hypoxia. ACS APPLIED NANO MATERIALS 2024; 7:10243-10256. [DOI: 10.1021/acsanm.4c00580] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Affiliation(s)
- Jieling Zheng
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou 510627, China
| | - Zicong He
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou 510627, China
| | - Luyan Shen
- Engineering Research Center for Nanophotonics and Advanced Instrument, School of Physics and Electronic Science, East China Normal University, Shanghai 200241, China
| | - Xiaoyu Chen
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Pei Chen
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou 510627, China
| | - Bin Zhang
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou 510627, China
| | - Huan Qin
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Zhiyuan Xiong
- School of Light Industry and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Shuixing Zhang
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou 510627, China
| |
Collapse
|
16
|
Augustin RC, Luke JJ. Rapidly Evolving Pre- and Post-surgical Systemic Treatment of Melanoma. Am J Clin Dermatol 2024; 25:421-434. [PMID: 38409643 PMCID: PMC11552441 DOI: 10.1007/s40257-024-00852-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2024] [Indexed: 02/28/2024]
Abstract
With the development of effective BRAF-targeted and immune-checkpoint immunotherapies for metastatic melanoma, clinical trials are moving these treatments into earlier adjuvant and perioperative settings. BRAF-targeted therapy is a standard of care in resected stage III-IV melanoma, while anti-programmed death-1 (PD1) immunotherapy is now a standard of care option in resected stage IIB through IV disease. With both modalities, recurrence-free survival and distant-metastasis-free survival are improved by a relative 35-50%, yet no improvement in overall survival has been demonstrated. Neoadjuvant anti-PD1 therapy improves event-free survival by approximately an absolute 23%, although improvements in overall survival have yet to be demonstrated. Understanding which patients are most likely to recur and which are most likely to benefit from treatment is now the highest priority question in the field. Biomarker analyses, such as gene expression profiling of the primary lesion and circulating DNA, are preliminarily exciting as potential biomarkers, though each has drawbacks. As in the setting of metastatic disease, markers that inform positive outcomes include interferon-γ gene expression, PD-L1, and high tumor mutational burden, while negative predictors of outcome include circulating factors such as lactate dehydrogenase, interleukin-8, and C-reactive protein. Integrating and validating these markers into clinically relevant models is thus a high priority. Melanoma therapeutics continues to advance with combination adjuvant approaches now investigating anti-PD1 with lymphocyte activation gene 3 (LAG3), T-cell immunoreceptor with Ig and ITIM domains (TIGIT), and individualized neoantigen therapies. How this progress will be integrated into the management of a unique patient to reduce recurrence, limit toxicity, and avoid over-treatment will dominate clinical research and patient care over the next decade.
Collapse
Affiliation(s)
- Ryan C Augustin
- UPMC Hillman Cancer Center, 5150 Centre Ave. Room 1.27C, Pittsburgh, PA, 15232, USA
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Jason J Luke
- UPMC Hillman Cancer Center, 5150 Centre Ave. Room 1.27C, Pittsburgh, PA, 15232, USA.
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
17
|
O'Rourke H, Hart C, De Boer RH. Current usage of pembrolizumab in triple negative breast cancer (TNBC). Expert Rev Anticancer Ther 2024; 24:253-261. [PMID: 38594892 DOI: 10.1080/14737140.2024.2341729] [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: 11/07/2023] [Accepted: 04/08/2024] [Indexed: 04/11/2024]
Abstract
INTRODUCTION The use of immune checkpoint inhibitors (ICI) targeting the PD-1/PD-L1 pathway has changed the landscape in the treatment of triple negative breast cancer (TNBC). The ICI pembrolizumab in combination with chemotherapy now forms a standard of care for the treatment of advanced PD-L1 positive TNBC and as part of neoadjuvant therapy for high-risk early-stage disease. Evidence in this space is rapidly advancing. AREAS COVERED This review aims to highlight the evolving role of immunotherapy in TNBC management and to discuss current challenges. The studies in this review were searched from PubMed and ClinicalTrials.gov. EXPERT OPINION The KEYNOTE-522 trial demonstrated that the addition of peri-operative pembrolizumab to neoadjuvant chemotherapy improves patient outcomes in early-stage TNBC. However, critical questions remain including how to select which patients truly gain benefit from the addition of pembrolizumab; the optimal duration of therapy, and the optimal adjuvant therapy depending on pathologic response.
Collapse
Affiliation(s)
- Harriet O'Rourke
- Department of Medical Oncology, St Vincent's Hospital, Fitzroy, Victoria, Australia
| | - Christopher Hart
- Department of Medical Oncology, St Vincent's Hospital, Fitzroy, Victoria, Australia
| | - Richard H De Boer
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| |
Collapse
|
18
|
Dabbaghipour R, Ahmadi E, Entezam M, Farzam OR, Sohrabi S, Jamali S, Sichani AS, Paydar H, Baradaran B. Concise review: The heterogenous roles of BATF3 in cancer oncogenesis and dendritic cells and T cells differentiation and function considering the importance of BATF3-dependent dendritic cells. Immunogenetics 2024; 76:75-91. [PMID: 38358555 DOI: 10.1007/s00251-024-01335-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 12/23/2023] [Indexed: 02/16/2024]
Abstract
The transcription factor, known as basic leucine zipper ATF-like 3 (BATF3), is a crucial contributor to the development of conventional type 1 dendritic cells (cDC1), which is definitely required for priming CD8 + T cell-mediated immunity against intracellular pathogens and malignancies. In this respect, BATF3-dependent cDC1 can bring about immunological tolerance, an autoimmune response, graft immunity, and defense against infectious agents such as viruses, microbes, parasites, and fungi. Moreover, the important function of cDC1 in stimulating CD8 + T cells creates an excellent opportunity to develop a highly effective target for vaccination against intracellular pathogens and diseases. BATF3 has been clarified to control the development of CD8α+ and CD103+ DCs. The presence of BATF3-dependent cDC1 in the tumor microenvironment (TME) reinforces immunosurveillance and improves immunotherapy approaches, which can be beneficial for cancer immunotherapy. Additionally, BATF3 acts as a transcriptional inhibitor of Treg development by decreasing the expression of the transcription factor FOXP3. However, when overexpressed in CD8 + T cells, it can enhance their survival and facilitate their transition to a memory state. BATF3 induces Th9 cell differentiation by binding to the IL-9 promoter through a BATF3/IRF4 complex. One of the latest research findings is the oncogenic function of BATF3, which has been approved and illustrated in several biological processes of proliferation and invasion.
Collapse
Affiliation(s)
- Reza Dabbaghipour
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Ahmadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mona Entezam
- Department of Medical Genetics, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Omid Rahbar Farzam
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepideh Sohrabi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sajjad Jamali
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Saber Sichani
- Department of Medical Genetics, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Biology, Texas A&M University, College Station, TX, 77843, USA
| | - Hadi Paydar
- Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
19
|
Middha P, Thummalapalli R, Betti MJ, Yao L, Quandt Z, Balaratnam K, Bejan CA, Cardenas E, Falcon CJ, Faleck DM, Gubens MA, Huntsman S, Johnson DB, Kachuri L, Khan K, Li M, Lovly CM, Murray MH, Patel D, Werking K, Xu Y, Zhan LJ, Balko JM, Liu G, Aldrich MC, Schoenfeld AJ, Ziv E. Polygenic risk score for ulcerative colitis predicts immune checkpoint inhibitor-mediated colitis. Nat Commun 2024; 15:2568. [PMID: 38531883 PMCID: PMC10966072 DOI: 10.1038/s41467-023-44512-4] [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: 05/22/2023] [Accepted: 12/15/2023] [Indexed: 03/28/2024] Open
Abstract
Immune checkpoint inhibitor-mediated colitis (IMC) is a common adverse event of treatment with immune checkpoint inhibitors (ICI). We hypothesize that genetic susceptibility to Crohn's disease (CD) and ulcerative colitis (UC) predisposes to IMC. In this study, we first develop a polygenic risk scores for CD (PRSCD) and UC (PRSUC) in cancer-free individuals and then test these PRSs on IMC in a cohort of 1316 patients with ICI-treated non-small cell lung cancer and perform a replication in 873 ICI-treated pan-cancer patients. In a meta-analysis, the PRSUC predicts all-grade IMC (ORmeta=1.35 per standard deviation [SD], 95% CI = 1.12-1.64, P = 2×10-03) and severe IMC (ORmeta=1.49 per SD, 95% CI = 1.18-1.88, P = 9×10-04). PRSCD is not associated with IMC. Furthermore, PRSUC predicts severe IMC among patients treated with combination ICIs (ORmeta=2.20 per SD, 95% CI = 1.07-4.53, P = 0.03). Overall, PRSUC can identify patients receiving ICI at risk of developing IMC and may be useful to monitor patients and improve patient outcomes.
Collapse
Affiliation(s)
- Pooja Middha
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Rohit Thummalapalli
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael J Betti
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lydia Yao
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Zoe Quandt
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
- Diabetes Center, University of California San Francisco, San Francisco, CA, USA
| | | | - Cosmin A Bejan
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Eduardo Cardenas
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Christina J Falcon
- Fiona and Stanley Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David M Faleck
- Gastroenterology, Hepatology & Nutrition Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Matthew A Gubens
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Scott Huntsman
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Douglas B Johnson
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Linda Kachuri
- Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cancer Institute, Stanford University of Medicine, Stanford, CA, USA
| | - Khaleeq Khan
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Min Li
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Christine M Lovly
- Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center, Nashville, TN, USA
| | - Megan H Murray
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Kristin Werking
- Department of Thoracic Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Yaomin Xu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Luna Jia Zhan
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Justin M Balko
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Geoffrey Liu
- Princess Margaret Cancer Centre, Toronto, ON, Canada
- Temerty School of Medicine, Toronto, ON, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Melinda C Aldrich
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Adam J Schoenfeld
- Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elad Ziv
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA.
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA.
- Center for Genes, Environment and Health, University of California San Francisco, San Francisco, CA, USA.
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
20
|
Zhou H, Yu CY, Wei H. Liposome-based nanomedicine for immune checkpoint blocking therapy and combinatory cancer therapy. Int J Pharm 2024; 652:123818. [PMID: 38253269 DOI: 10.1016/j.ijpharm.2024.123818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 01/06/2024] [Accepted: 01/16/2024] [Indexed: 01/24/2024]
Abstract
The discovery of immune checkpoint (IC) has led to a wave of leap forward in cancer immunotherapy that represents probably the most promising strategy for cancer therapy. However, the clinical use of immune checkpoint block (ICB) therapy is limited by response rates and side effects. A strategy that addresses the limitations of ICB therapies through combination therapies, using nanocarriers as mediators, has been mentioned in numerous research papers. Liposomes have been probably one of the most extensively used nanocarriers for clinical applications, with broad drug delivery and high safety. A timely review on this hot subject of research, i.e., the application of liposomes for ICB, is thus highly desirable for both fundamental and clinical translatable studies, but remains, to our knowledge, unexplored so far. For this purpose, this review is composed to address the dilemma of ICB therapy and the reasons for this dilemma. We later describe how other cancer treatments have broken this dilemma. Finally, we focus on the role of liposomes in various combinatory cancer therapy. This review is believed to serve as a guidance for the rational design and development of liposome for immunotherapy with enhanced therapeutic efficiency.
Collapse
Affiliation(s)
- Haoyuan Zhou
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical of Science, Hengyang 421001, China
| | - Cui-Yun Yu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical of Science, Hengyang 421001, China.
| | - Hua Wei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical of Science, Hengyang 421001, China.
| |
Collapse
|
21
|
O’Meara CH, Nguyen TV, Jafri Z, Boyer M, Shonka DC, Khachigian LM. Personalised Medicine and the Potential Role of Electrospinning for Targeted Immunotherapeutics in Head and Neck Cancer. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 14:6. [PMID: 38202461 PMCID: PMC10780990 DOI: 10.3390/nano14010006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 12/03/2023] [Accepted: 12/15/2023] [Indexed: 01/12/2024]
Abstract
Advanced head and neck cancer (HNC) is functionally and aesthetically destructive, and despite significant advances in therapy, overall survival is poor, financial toxicity is high, and treatment commonly exacerbates tissue damage. Although response and durability concerns remain, antibody-based immunotherapies have heralded a paradigm shift in systemic treatment. To overcome limitations associated with antibody-based immunotherapies, exploration into de novo and repurposed small molecule immunotherapies is expanding at a rapid rate. Small molecule immunotherapies also have the capacity for chelation to biodegradable, bioadherent, electrospun scaffolds. This article focuses on the novel concept of targeted, sustained release immunotherapies and their potential to improve outcomes in poorly accessible and risk for positive margin HNC cases.
Collapse
Affiliation(s)
- Connor H. O’Meara
- Department of Otorhinolaryngology, Head & Neck Surgery, The Canberra Hospital, Garran, ACT 2605, Australia
- ANU School of Medicine, Australian National University, Canberra, ACT 0200, Australia
| | - Thanh Vinh Nguyen
- School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia;
| | - Zuhayr Jafri
- Vascular Biology and Translational Research, Department of Pathology, School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia; (Z.J.)
| | - Michael Boyer
- Chris O’Brien Lifehouse, Camperdown, NSW 2050, Australia;
| | - David C. Shonka
- Department of Otolaryngology, Head & Neck Surgery, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - Levon M. Khachigian
- Vascular Biology and Translational Research, Department of Pathology, School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia; (Z.J.)
| |
Collapse
|
22
|
Guo Z, Cai Y, Yin W, Huang J. A locally advanced colon cancer patient with Muir-Torre syndrome obtains durable response to neoadjuvant and adjuvant immunotherapy. TUMORI JOURNAL 2023; 109:NP27-NP31. [PMID: 37880978 DOI: 10.1177/03008916231204735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2023]
Abstract
INTRODUCTION Muir-Torre syndrome, presenting with cutaneous tumors and visceral malignancies, is a variant of Lynch syndrome. The development of immune checkpoint inhibitors provided novel effective treatment options for metastatic colorectal cancer patients with microsatellite instability and deficient mismatch repair. However, the use of immune checkpoint inhibitors in neoadjuvant and adjuvant settings for patients with locally advanced colorectal cancer remains undefined because of limited follow-ups in current studies. CASE PRESENTATION In the present study, we reported a 33-year-old Muri-Torre syndrome patient with stage ⅢC (c.T4N2M0) colorectal cancer and keratoacanthoma. Microsatellite instability / deficient mismatch repair, high tumor mutation burden, and MSH2 germline mutation were identified by next-generation sequencing. Pembrolizumab monotherapy was used as neoadjuvant treatment and the patient achieved a major pathological response. After surgical resection, pembrolizumab was continuously used in an adjuvant setting for 12 months. The patient remained disease-free with a durable disease-free survival for 44 months. To our knowledge, this is the first and longest follow-up study reporting pembrolizumab as a single-agent neoadjuvant therapy for locally advanced colon cancer. CONCLUSIONS The results demonstrate promising performance in neoadjuvant and adjuvant settings. Further studies are needed to confirm its potential usefulness as an outcome measure in clinical practice.
Collapse
Affiliation(s)
- Zhihua Guo
- Department of Thoracic Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Institute of Respiratory Disease and State Key Laboratory for Respiratory Disease, Guangzhou, China
- Research Center for Translational Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yue Cai
- Department of Medical Oncology, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Weiqiang Yin
- Department of Thoracic Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Institute of Respiratory Disease and State Key Laboratory for Respiratory Disease, Guangzhou, China
- Research Center for Translational Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jun Huang
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
23
|
Blaise M, Poissonnet G, Cardot-Leccia N, Sudaka A, Velin M, Chassang M, Razzouk-Cadet M, Picard-Gauci A, Passeron T, Montaudié H. Pathological complete response to neoadjuvant pembrolizumab in a patient with metastatic cutaneous squamous cell carcinoma. J Eur Acad Dermatol Venereol 2023; 37:e1318-e1320. [PMID: 37415448 DOI: 10.1111/jdv.19307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 06/27/2023] [Indexed: 07/08/2023]
Affiliation(s)
- Manon Blaise
- Dermatology Department, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France
| | - Gilles Poissonnet
- Cervico-facial Department, Institut Universitaire de la Face et du Cou, Nice, France
| | - Nathalie Cardot-Leccia
- Histopathology Department, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France
| | - Anne Sudaka
- Histopathology Department, Centre Antoine Lacassagne, Nice, France
| | - Marine Velin
- Dermatology Department, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France
| | - Madleen Chassang
- Radiology Department, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France
| | - Micheline Razzouk-Cadet
- Nuclear Medicine Department, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France
| | - Alexandra Picard-Gauci
- Dermatology Department, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France
| | - Thierry Passeron
- Dermatology Department, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France
- INSERM U1065, C3M, Université Côte d'Azur, Nice, France
| | - Henri Montaudié
- Dermatology Department, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France
- INSERM U1065, C3M, Université Côte d'Azur, Nice, France
| |
Collapse
|
24
|
Attieh F, Chartouni A, Boutros M, Mouawad A, Kourie HR. Tackling the immunotherapy conundrum: advances and challenges for operable non-small-cell lung cancer treatment. Immunotherapy 2023; 15:1415-1428. [PMID: 37671552 DOI: 10.2217/imt-2023-0128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2023] Open
Abstract
Lung cancer is the most common cause of cancer-related deaths worldwide. Non-small-cell lung cancer (NSCLC) represents the majority of lung cancer cases, and its standard treatment is primarily surgery. Nonetheless, this type of cancer exhibits an important rate of tumor recurrence. Immune checkpoint inhibitors (ICIs) have demonstrated significant survival benefits in many cancers, especially in early-stage NSCLC. This review considers the latest CheckMate816, IMpower010 and KEYNOTE-091 trials that led to US FDA approvals. The new wave of resectable NSCLC trial results are also summarized. Finally, the latest challenges for these treatment modalities, such as the choice between neoadjuvant and adjuvant use, the accurate identification of biomarkers and the presence of driver mutations such as EGFR, are discussed.
Collapse
Affiliation(s)
- Fouad Attieh
- Faculty of Medicine, Saint Joseph University of Beirut, Beirut, 11072180, Lebanon
| | - Antoine Chartouni
- Faculty of Medicine, Saint Joseph University of Beirut, Beirut, 11072180, Lebanon
| | - Marc Boutros
- Faculty of Medicine, Saint Joseph University of Beirut, Beirut, 11072180, Lebanon
| | - Antoine Mouawad
- Faculty of Medicine, Saint Joseph University of Beirut, Beirut, 11072180, Lebanon
| | - Hampig Raphaël Kourie
- Department of Hematology-Oncology, Faculty of Medicine, Saint Joseph University of Beirut, Beirut, 11072180, Lebanon
| |
Collapse
|
25
|
Liang Q, Chen J, Hou S, Li D, Zhu Y, Li R, Chen L, Li J, Fu W, Lei S, Zhang B, Zheng X, Zhang T, Duan H, He W, Ren J. Activatable Mn 2+-Armed nanoagonist augments antitumor immunity in colorectal cancer: A NIR-II Photonic neoadjuvant paradigm. Biomaterials 2023; 300:122206. [PMID: 37348325 DOI: 10.1016/j.biomaterials.2023.122206] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 05/24/2023] [Accepted: 06/11/2023] [Indexed: 06/24/2023]
Abstract
Postoperative recurrence frequently occurs in patients with colorectal cancer (CRC) due to residual microtumors and host cellular immune dysfunction, leading to major setbacks in clinical outcomes and CRC staging. As an increasingly prevalent therapeutic option for CRC patients, neoadjuvant chemoradiotherapy bears unmet challenges of limited tumor targeting and common side effects of gastrointestinal reaction and radiodermatitis. It is highly desirable to develop neoadjuvant treatment paradigms that impart both tumor-targeting accuracy and protection against recurrence of resectable CRC. Here we report a versatile photo-regulated nanoagonist of plasmonic gold blackbody (AuPB) with a polydopamine (PDA) coating carrying manganese ion (Mn2+) payloads (AuPB@PDA/Mn). When armed with second near-infrared (NIR-II) light, AuPB@PDA/Mn with broad-band localized surface plasmon resonance generates local hyperthermia and discharges Mn2+ ions, which evidently amplify the effects of immunogenic cell death in tumor cells and activate the cyclic GMP-AMP synthase/stimulator of interferon genes pathway in dendritic cells (DCs), hence potentiating the maturation of DC and the secretion of type I interferon in a synergistic way. Matured DCs undertake the task of tumor antigen presentation as the crosstalk to adaptive immunity. As such, the administration of AuPB@PDA/Mn coupled with NIR-II laser irradiation has eminently augmented the infiltration of CD8+ T cells as well as the development of memory CD8+ T cells in colorectal tumor models, substantiating enhanced immunomodulatory efficacy against primary and recurrent CRC. Our strategy highlights the potency of an integrated NIR-II photothermal and immunoregulatory modality by photo-activate delivery of Mn2+ ions, as a neoadjuvant paradigm for presurgical tumor debulking and against postoperative tumor recurrence.
Collapse
Affiliation(s)
- Qing Liang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jiayuan Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Shuai Hou
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Di Li
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Ying Zhu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ruiqi Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Lian Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jiao Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wei Fu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Shiqiong Lei
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Biying Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xin Zheng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Tao Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hongwei Duan
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore.
| | - Wenshan He
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Jinghua Ren
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
26
|
Walker PR. Induction treatment in operable non-small cell lung cancer: the immune oncology era unfolds. J Thorac Dis 2023; 15:3533-3537. [PMID: 37559643 PMCID: PMC10407505 DOI: 10.21037/jtd-23-699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 06/02/2023] [Indexed: 08/11/2023]
|
27
|
Bushara O, Tidwell J, Wester JR, Miura J. The Current State of Neoadjuvant Therapy in Resectable Advanced Stage Melanoma. Cancers (Basel) 2023; 15:3344. [PMID: 37444454 DOI: 10.3390/cancers15133344] [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: 04/22/2023] [Revised: 06/17/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
The advent of effective immunotherapy and targeted therapy has significantly improved outcomes in advanced-stage resectable melanoma. Currently, the mainstay of treatment of malignant melanoma is surgery followed by adjuvant systemic therapies. However, recent studies have shown a potential role for neoadjuvant therapy in the treatment of advanced-stage resectable melanoma. Mechanistically, neoadjuvant immunotherapy may yield a more robust response than adjuvant immunotherapy, as the primary tumor serves as an antigen in this setting rather than only micrometastatic disease after the index procedure. Additionally, targeted therapy has been shown to yield effective neoadjuvant cytoreduction, and oncolytic viruses may also increase the immunogenicity of primary tumors. Effective neoadjuvant therapy may serve to decrease tumor size and thus reduce the extent of required surgery and thus morbidity. It also allows for assessment of pathologic response, facilitating prognostication as well as tailoring future therapy. The current literature consistently supports that neoadjuvant therapy, even as little as one dose, is associated with improved outcomes and is well-tolerated. Some patients with a complete pathological response may even avoid surgery completely. These results challenge the current paradigm of a surgery-first approach and provide further evidence supporting neoadjuvant therapy in advanced-stage resectable melanoma. Further research into the optimal treatment schedule and dose timing is warranted, as is the continued investigation of novel therapies and combinations of therapies.
Collapse
Affiliation(s)
- Omar Bushara
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jerica Tidwell
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - James R Wester
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - John Miura
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
28
|
Nindra U, Hurwitz J, Forstner D, Chin V, Gallagher R, Liu J. A systematic review of neoadjuvant and definitive immunotherapy in locally advanced head and neck squamous cell carcinoma. Cancer Med 2023. [PMID: 36934434 DOI: 10.1002/cam4.5815] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 02/28/2023] [Accepted: 03/06/2023] [Indexed: 03/20/2023] Open
Abstract
BACKGROUND Patients with locally advanced head and neck squamous cell carcinoma (HNSCC) require multi-modality treatment. Immune checkpoint inhibitors (ICIs) are now standard of care in management of recurrent/metastatic HNSCC. However, its role in the definitive and neoadjuvant setting remains unclear. METHODS A literature search was conducted that included all articles investigating ICI in untreated locally advanced (LA) HNSCC. Data was extracted and summarised and rated for quality using the Cochrane risk of bias tool. RESULTS Of 1086 records, 29 met the final inclusion criteria. In both concurrent and neoadjuvant settings, the addition of ICI was safe and did not delay surgery or reduce chemoradiotherapy completion. In the concurrent setting, although ICI use demonstrates objective responses in all published trials, there has not yet been published data to with PFS or OS benefit. In the neoadjuvant setting, combination ICI resulted in superior major pathological response rates compared to ICI monotherapy without a significant increase adverse event profiles, but its value in improving survival is not clear. ICI efficacy appears to be affected by tumour characteristics, in particular PD-L1 combined positive score, HPV status and the tumour microenvironment. CONCLUSIONS There is significant heterogeneity of ICI use in untreated LA HNSCC with multiple definitive concurrent and neoadjuvant protocols used. Resultantly, conclusions regarding the survival benefits of adding ICI to standard-of-care regimens cannot be made. Further trials and translational studies are required to elucidate optimal ICI sequencing in the definitive setting as well as better define populations more suited for neoadjuvant protocols.
Collapse
Affiliation(s)
- Udit Nindra
- Department of Medical Oncology, Liverpool Hospital, Sydney, New South Wales, Australia.,Department of Medical Oncology, Campbelltown Hospital, Sydney, New South Wales, Australia
| | - Joshua Hurwitz
- The Kinghorn Cancer Centre, St Vincent's Hospital, Darlinghurst, New South Wales, Australia
| | - Dion Forstner
- The Kinghorn Cancer Centre, St Vincent's Hospital, Darlinghurst, New South Wales, Australia.,The University of New South Wales, Kensington, New South Wales, Australia.,GenesisCare, Darlinghurst, New South Wales, Australia
| | - Venessa Chin
- The Kinghorn Cancer Centre, St Vincent's Hospital, Darlinghurst, New South Wales, Australia.,The University of New South Wales, Kensington, New South Wales, Australia.,The Garvan Institute of Research, Camperdown, New South Wales, Australia
| | - Richard Gallagher
- The Kinghorn Cancer Centre, St Vincent's Hospital, Darlinghurst, New South Wales, Australia.,The University of New South Wales, Kensington, New South Wales, Australia.,The University of Sydney, Camperdown, New South Wales, Australia
| | - Jia Liu
- The Kinghorn Cancer Centre, St Vincent's Hospital, Darlinghurst, New South Wales, Australia.,The University of New South Wales, Kensington, New South Wales, Australia.,The University of Sydney, Camperdown, New South Wales, Australia
| |
Collapse
|
29
|
Wang P, Liu X, Yu J, Meng Z, Lv Z, Shang C, Geng Q, Wang D, Xue D, Li L. Fucosyltransferases Regulated by Fusobacterium Nucleatum and Act as Novel Biomarkers in Colon Adenocarcinoma. J Inflamm Res 2023; 16:747-768. [PMID: 36852302 PMCID: PMC9960735 DOI: 10.2147/jir.s396484] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 02/03/2023] [Indexed: 02/22/2023] Open
Abstract
Purpose Colon adenocarcinoma (COAD) is one of the leading causes of cancer-associated mortality worldwide. Fucosyltransferases (FUTs) are associated with numerous cancers. We aimed to investigate the functions of FUTs in COAD. Patients and Methods Transcriptomic and clinical data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases were used to analyze the expression and clinical relevance of FUTs in COAD. Real Time Quantitative PCR (RT-qPCR), Western blot, immunohistochemistry and ELISA were used to detect the relative RNA and protein expression levels. Colitis-associated cancer mice treated with Fusobacterium nucleatum were used to illustrate the effects of Fusobacterium nucleatum on FUTs and COAD. Luciferase reporting assay was used to investigate the binding of miRNA to mRNA. Results TCGA and GEO datasets showed abnormal expression of FUTs in COAD at transcript level. RT-qPCR, Western blot and immunohistochemistry showed increased expression of FUT1, POFUT1 and POFUT2 in COAD. COAD patients with a high expression of FUT1, FUT11, FUT13 (POFUT2) had a worse prognosis, while patients with a high expression of FUT2, FUT3, FUT6 had a better prognosis. FUT1 and POFUT2 could independently predict the prognosis of COAD patients. Functional analysis by CancerSEA database showed that FUT3, FUT6, FUT8, FUT12 (POFUT1) and FUT13 are associated with differentiation, apoptosis, invasion, quiescence, and hypoxia. FUTs are associated with the tumor microenvironment of COAD. FUT1 regulated by miR-939-3p inhibit the expression of MUC2. Fusobacterium nucleatum may affect the expression of FUTs by affecting their transcription factors and miRNA levels. Moreover, Fusobacterium nucleatum promotes COAD progression through the miR-939-3p/FUT1/MUC2 axis. Conclusion Fucosyltransferases play an important role and may be the mediator of Fusobacterium nucleatum promoting COAD progression.
Collapse
Affiliation(s)
- Pengfei Wang
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Xuxu Liu
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Jingjing Yu
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Ziang Meng
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Zhenyi Lv
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Ce Shang
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Qi Geng
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Dawei Wang
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China,Correspondence: Dawei Wang, Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin, 150001, People’s Republic of China, Tel/Fax +86 451 85555776, Email
| | - Dongbo Xue
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Long Li
- Intestinal Microenvironment Treatment Center of General Surgery, Shanghai Tenth People’s Hospital, Tenth People’s Hospital of Tongji University, Shanghai, People’s Republic of China,Long Li, Intestinal Microenvironment Treatment Center of General Surgery, Shanghai Tenth People’s Hospital, Tenth People’s Hospital of Tongji University, 301 Yanchang Middle Road, Shanghai, 200072, People’s Republic of China, Tel/Fax +86 21 66307011, Email
| |
Collapse
|
30
|
Lampridis S, Scarci M. Perioperative systemic therapies for non-small-cell lung cancer: Recent advances and future perspectives. Front Surg 2023; 9:1126486. [PMID: 36743902 PMCID: PMC9895369 DOI: 10.3389/fsurg.2022.1126486] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 12/31/2022] [Indexed: 01/22/2023] Open
Abstract
The mainstay of treatment for early-stage non-small-cell lung cancer (NSCLC) is surgical resection. Traditionally, chemotherapy has been used perioperatively in locally extensive disease to improve the oncologic outcomes of surgery, with a 5-year absolute survival benefit of approximately 5%. In recent years, immunotherapy and molecular targeted therapy have shown excellent results in the treatment of locoregionally advanced and metastatic NSCLC, replacing chemotherapy as first-line treatment in certain cases. Consequently, researchers have been increasingly investigating the use of immunotherapy or targeted therapy in combination with surgery for the treatment of early-stage disease. This growing research interest has resulted in several published and ongoing studies of various size and design. In this mini review, we provide a succinct and up-to-date overview of recently published, phase 3 randomized clinical trials on adjuvant and neoadjuvant immunotherapy or targeted therapy for NSCLC. We subsequently discuss some important unresolved clinical issues, including the optimal duration of treatment, scheduling with respect to surgery, and potential combinations of different systemic therapies. Finally, we reference large, randomized, phase 3 studies that are currently in progress and may give answers to those and other clinical questions.
Collapse
Affiliation(s)
- Savvas Lampridis
- Department of Cardiothoracic Surgery, Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom
| | | |
Collapse
|
31
|
Ouranos K, Chatziioannou A, Goulis I, Sinakos E. Role of immunotherapy in downsizing hepatocellular carcinoma prior to liver transplantation. World J Transplant 2022; 12:331-346. [PMID: 36437845 PMCID: PMC9693898 DOI: 10.5500/wjt.v12.i11.331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/10/2022] [Accepted: 10/19/2022] [Indexed: 11/17/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is an aggressive primary liver neoplasm that, according to tumor stage, can be treated with resection, transplantation, locoregional treatment options, or systemic therapy. Although interventions only in early-stage disease can offer complete tumor regression, systemic therapy in advanced disease can significantly prolong overall survival, according to published clinical trials. The emergence of immunotherapy in the field of cancer therapy has had a positive impact on patients with HCC, resulting in atezolizumab–bevacizumab currently being the first-line option for treatment of advanced HCC. In light of this, application of immunotherapy in the preoperative process could increase the number of patients fulfilling the criteria for liver transplantation (LT). Implementation of this approach is faced with challenges regarding the safety of immunotherapy and the possibly increased risk of rejection in the perioperative period. Case reports and clinical trials assessing the safety profile and effectiveness of neoadjuvant immunotherapy, highlight important aspects regarding this newly evolving approach to HCC management. More studies need to be conducted in order to reach a consensus regarding the optimal way to administer immunotherapy prior to LT. In this review, we summarize the role, safety profile and future considerations regarding the use of neoadjuvant immunotherapy prior to LT in patients with HCC.
Collapse
Affiliation(s)
- Konstantinos Ouranos
- 4th Medical Department, Hippokratio Hospital, Aristotle University of Thessaloniki, Thessaloniki 54642, Greece
| | - Anthi Chatziioannou
- 4th Medical Department, Hippokratio Hospital, Aristotle University of Thessaloniki, Thessaloniki 54642, Greece
| | - Ioannis Goulis
- 4th Medical Department, Hippokratio Hospital, Aristotle University of Thessaloniki, Thessaloniki 54642, Greece
| | - Emmanouil Sinakos
- 4th Medical Department, Hippokratio Hospital, Aristotle University of Thessaloniki, Thessaloniki 54642, Greece
| |
Collapse
|
32
|
Cheng X, Zhang H, Hamad A, Huang H, Tsung A. Surgery-mediated tumor-promoting effects on the immune microenvironment. Semin Cancer Biol 2022; 86:408-419. [PMID: 35066156 PMCID: PMC11770836 DOI: 10.1016/j.semcancer.2022.01.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 02/07/2023]
Abstract
Surgical resection continues to be the mainstay treatment for solid cancers even though chemotherapy and immunotherapy have significantly improved patient overall survival and progression-free survival. Numerous studies have shown that surgery induces the dissemination of circulating tumor cells (CTCs) and that the resultant inflammatory response promotes occult tumor growth and the metastatic process by forming a supportive tumor microenvironment (TME). Surgery-induced platelet activation is one of the initial responses to a wound and the formation of fibrin clots can provide the scaffold for recruited inflammatory cells. Activated platelets can also shield CTCs to protect them from blood shear forces and promote CTCs evasion of immune destruction. Similarly, neutrophils are recruited to the fibrin clot and enhance cancer metastatic dissemination and progression by forming neutrophil extracellular traps (NETs). Activated macrophages are also recruited to surgical sites to facilitate the metastatic spread. More importantly, the body's response to surgical insult results in the recruitment and expansion of immunosuppressive cell populations (i.e. myeloid-derived suppressor cells and regulatory T cells) and in the suppression of natural killer (NK) cells that contribute to postoperative cancer recurrence and metastasis. In this review, we seek to provide an overview of the pro-tumorigenic mechanisms resulting from surgery's impact on these cells in the TME. Further understanding of these events will allow for the development of perioperative therapeutic strategies to prevent surgery-associated metastasis.
Collapse
Affiliation(s)
- Xiang Cheng
- Division of Surgical Oncology, Department of Surgery, The Ohio State University James Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Hongji Zhang
- Division of Surgical Oncology, Department of Surgery, The Ohio State University James Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Ahmad Hamad
- Division of Surgical Oncology, Department of Surgery, The Ohio State University James Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Hai Huang
- Division of Surgical Oncology, Department of Surgery, The Ohio State University James Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Allan Tsung
- Division of Surgical Oncology, Department of Surgery, The Ohio State University James Comprehensive Cancer Center, Columbus, OH, 43210, USA.
| |
Collapse
|
33
|
Atkins MB, Abu-Sbeih H, Ascierto PA, Bishop MR, Chen DS, Dhodapkar M, Emens LA, Ernstoff MS, Ferris RL, Greten TF, Gulley JL, Herbst RS, Humphrey RW, Larkin J, Margolin KA, Mazzarella L, Ramalingam SS, Regan MM, Rini BI, Sznol M. Maximizing the value of phase III trials in immuno-oncology: A checklist from the Society for Immunotherapy of Cancer (SITC). J Immunother Cancer 2022; 10:jitc-2022-005413. [PMID: 36175037 PMCID: PMC9528604 DOI: 10.1136/jitc-2022-005413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2022] [Indexed: 11/03/2022] Open
Abstract
The broad activity of agents blocking the programmed cell death protein 1 and its ligand (the PD-(L)1 axis) revolutionized oncology, offering long-term benefit to patients and even curative responses for tumors that were once associated with dismal prognosis. However, only a minority of patients experience durable clinical benefit with immune checkpoint inhibitor monotherapy in most disease settings. Spurred by preclinical and correlative studies to understand mechanisms of non-response to the PD-(L)1 antagonists and by combination studies in animal tumor models, many drug development programs were designed to combine anti-PD-(L)1 with a variety of approved and investigational chemotherapies, tumor-targeted therapies, antiangiogenic therapies, and other immunotherapies. Several immunotherapy combinations improved survival outcomes in a variety of indications including melanoma, lung, kidney, and liver cancer, among others. This immunotherapy renaissance, however, has led to many combinations being advanced to late-stage development without definitive predictive biomarkers, limited phase I and phase II data, or clinical trial designs that are not optimized for demonstrating the unique attributes of immune-related antitumor activity-for example, landmark progression-free survival and overall survival. The decision to activate a study at an individual site is investigator-driven, and generalized frameworks to evaluate the potential for phase III trials in immuno-oncology to yield positive data, particularly to increase the number of curative responses or otherwise advance the field have thus far been lacking. To assist in evaluating the potential value to patients and the immunotherapy field of phase III trials, the Society for Immunotherapy of Cancer (SITC) has developed a checklist for investigators, described in this manuscript. Although the checklist focuses on anti-PD-(L)1-based combinations, it may be applied to any regimen in which immune modulation is an important component of the antitumor effect.
Collapse
Affiliation(s)
- Michael B Atkins
- Georgetown Lombardi Comprehensive Cancer Center, Washington, District of Columbia, USA
| | | | - Paolo A Ascierto
- Istituto Nazionale Tumori IRCCS Fondazione "G Pascale", Napoli, Italy
| | - Michael R Bishop
- The David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, Illinois, USA
| | - Daniel S Chen
- Engenuity Life Sciences, Burlingame, California, USA
| | - Madhav Dhodapkar
- Center for Cancer Immunology, Winship Cancer Institute at Emory University, Atlanta, Georgia, USA
| | - Leisha A Emens
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Marc S Ernstoff
- DCTD/DTP-IOB, ImmunoOncology Branch, NCI, Bethesda, Maryland, USA
| | | | - Tim F Greten
- Gastrointestinal Malignancies Section, National Cancer Institue CCR Liver Program, Bethesda, Maryland, USA
| | - James L Gulley
- Center for Immuno-Oncology, National Cancer Institute, Bethesda, Maryland, USA
| | | | | | | | - Kim A Margolin
- St. John's Cancer Institute, Santa Monica, California, USA
| | - Luca Mazzarella
- Experimental Oncology, New Drug Development, European Instititue of Oncology IRCCS, Milan, Italy
| | | | - Meredith M Regan
- Dana-Farber/Harvard Cancer Center, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | | | - Mario Sznol
- Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
34
|
Igaue S, Okuno T, Ishibashi H, Nemoto M, Hiyoshi M, Kawasaki H, Saitoh H, Saitoh M, Akagi K, Yamamoto J. A pathological complete response after nivolumab plus ipilimumab therapy for DNA mismatch repair‑deficient/microsatellite instability‑high metastatic colon cancer: A case report. Oncol Lett 2022; 24:211. [PMID: 35720492 PMCID: PMC9178690 DOI: 10.3892/ol.2022.13332] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 04/28/2022] [Indexed: 12/24/2022] Open
Abstract
The standard treatment for colorectal cancer has always been surgery and chemotherapy, which may be used in combination to treat patients. Immune checkpoint inhibitors have been a significant advancement in the standard treatment of metastatic, unresectable colorectal cancer with deficient mismatch repair. However, little information is available about their use in neoadjuvant and conversion settings with only a few case reports and only one phase 2 trial. The present study reports the case of a large, locally advanced right-sided metastatic deficient mismatch repair/microsatellite instability-high colon cancer, which showed a pathological complete response after combination treatment with nivolumab and ipilimumab. To the best of our knowledge, resected metastatic colon cancer with a pathological complete response after treatment using dual immune checkpoint inhibitors has not been previously reported. Overall, this case report suggests the use of immune checkpoint inhibitors before colorectal surgery.
Collapse
Affiliation(s)
- Shota Igaue
- Department of Gastrointestinal Surgery, Ibaraki Cancer Center, Ibaraki Prefectural Central Hospital, Kasama, Ibaraki 309‑1793, Japan
| | - Takayuki Okuno
- Department of Gastrointestinal Surgery, Ibaraki Cancer Center, Ibaraki Prefectural Central Hospital, Kasama, Ibaraki 309‑1793, Japan
| | - Hajime Ishibashi
- Department of Gastroenterology, Ibaraki Cancer Center, Ibaraki Prefectural Central Hospital, Kasama, Ibaraki 309‑1793, Japan
| | - Masaru Nemoto
- Department of Gastrointestinal Surgery, Ibaraki Cancer Center, Ibaraki Prefectural Central Hospital, Kasama, Ibaraki 309‑1793, Japan
| | - Masaya Hiyoshi
- Department of Gastrointestinal Surgery, Ibaraki Cancer Center, Ibaraki Prefectural Central Hospital, Kasama, Ibaraki 309‑1793, Japan
| | - Hiroshi Kawasaki
- Department of Gastrointestinal Surgery, Ibaraki Cancer Center, Ibaraki Prefectural Central Hospital, Kasama, Ibaraki 309‑1793, Japan
| | - Hitoaki Saitoh
- Department of Pathology, Ibaraki Cancer Center, Ibaraki Prefectural Central Hospital, Kasama, Ibaraki 309‑1793, Japan
| | - Makoto Saitoh
- Center for Clinical Genetics and Genomics, Ibaraki Cancer Center, Ibaraki Prefectural Central Hospital, Kasama, Ibaraki 309‑1793, Japan
| | - Kiwamu Akagi
- Division of Molecular Diagnosis and Cancer Prevention, Saitama Cancer Center, Kitaadachi‑gun, Saitama 362‑0806, Japan
| | - Junji Yamamoto
- Department of Gastrointestinal Surgery, Ibaraki Cancer Center, Ibaraki Prefectural Central Hospital, Kasama, Ibaraki 309‑1793, Japan
| |
Collapse
|
35
|
Bilusic M. What are the advantages of neoadjuvant immunotherapy over adjuvant immunotherapy? Expert Rev Anticancer Ther 2022; 22:561-563. [PMID: 35473572 DOI: 10.1080/14737140.2022.2069097] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Marijo Bilusic
- Medical Oncology GU Site Disease Group Lead, Sylvester Comprehensive Cancer Center, University of Miami Health System, Miami, FL, USA
| |
Collapse
|
36
|
Agostinetto E, Losurdo A, Nader-Marta G, Santoro A, Punie K, Barroso R, Popovic L, Solinas C, Kok M, de Azambuja V, Lambertini M. Progress and pitfalls in the use of immunotherapy for patients with triple negative breast cancer. Expert Opin Investig Drugs 2022; 31:567-591. [PMID: 35240902 DOI: 10.1080/13543784.2022.2049232] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Triple negative breast cancer (TNBC) is an area of high unmet medical need in terms of new effective treatment strategies. Although breast cancer is traditionally considered a 'cold' tumor type, TNBC is the most appropriate subtype for immunotherapeutic strategies; this is due to the high level of tumor infiltrating lymphocytes, PD-L1 expression and tumor mutational burden relative to other breast cancer subtypes. AREAS COVERED This review examines the use of immunotherapeutic strategies in early and advanced TNBC. The paper summarizes data on novel promising immunomodulatory approaches that have been explored in early phase trials and discusses the pitfalls and limitations often encountered in clinical research. EXPERT OPINION PD-1-blockade is approved for stage II/III TNBC and for first-line treatment of PD-L1-positive TNBC patients with metastatic disease and should be considered standard of care. However, question marks and difficulties remain; these include the identification of predictive biomarkers to select patients who benefit from the addition of PD1-blockade and the balance between efficacy and long-term toxicity for an individual patient. Numerous treatment combinations and new immunotherapeutic strategies beyond PD1 blockade are being evaluated, thus reflecting a promising evolution of a more personalized approach, and extended clinical benefit in TNBC.
Collapse
Affiliation(s)
- Elisa Agostinetto
- Academic Trials Promoting Team, Institut Jules Bordet and l'Université Libre de Bruxelles (U.L.B), Brussels, Belgium.,Department of Biomedical Sciences, Humanitas University, via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Milan, Italy
| | - Agnese Losurdo
- Humanitas Research Hospital - IRCCS, Humanitas Cancer Center, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Guilherme Nader-Marta
- Academic Trials Promoting Team, Institut Jules Bordet and l'Université Libre de Bruxelles (U.L.B), Brussels, Belgium
| | - Armando Santoro
- Department of Biomedical Sciences, Humanitas University, via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Milan, Italy.,Humanitas Research Hospital - IRCCS, Humanitas Cancer Center, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Kevin Punie
- Department of General Medical Oncology and Multidisciplinary Breast Centre, Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium
| | | | - Lazar Popovic
- Oncology Institute of Vojvodina, Faculty of Medicine, University Novi Sad, Novi Sad, Serbia
| | - Cinzia Solinas
- Medical Oncology, ATS Sardegna, Ospedale San Francesco, Nuoro, Italy
| | - Marleen Kok
- Departments of Medical Oncology, Tumor Biology & Immunology. Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Vandro de Azambuja
- Academic Trials Promoting Team, Institut Jules Bordet and l'Université Libre de Bruxelles (U.L.B), Brussels, Belgium
| | - Matteo Lambertini
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy.,Department of Medical Oncology, U.O.C Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
37
|
Erstad DJ, Witt RG, Wargo JA. Neoadjuvant therapy for melanoma: new and evolving concepts. CLINICAL ADVANCES IN HEMATOLOGY & ONCOLOGY : H&O 2022; 20:47-55. [PMID: 35060962 PMCID: PMC9103060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Effective systemic therapies, including targeted BRAF/MEK inhibition and immune checkpoint blockade, have significantly changed the treatment landscape for malignant melanoma. Specifically, there have been promising clinical trial findings associated with the use of neoadjuvant therapy for clinically node-positive and oligometastatic disease, conditions that have historically been managed with up-front surgical resection when possible. This review focuses on the burgeoning field of neoadjuvant therapy for melanoma. We review the rationale for this treatment approach, summarize completed and ongoing neoadjuvant clinical trials, and contextualize these findings within the growing body of knowledge about targeted and immune checkpoint therapy. Finally, we discuss future directions for neoadjuvant trials in melanoma, with particular focus on biomarker development, treatment effect modification, novel therapeutic regimens, and evolving surgical indications for regional and oligometastatic disease.
Collapse
Affiliation(s)
- Derek J. Erstad
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Russell G. Witt
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jennifer A. Wargo
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA,Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
38
|
Mukherjee S, Parmar K, Smyth E. Immune checkpoint inhibitors in resectable gastroesophageal cancers - a review. Ther Adv Med Oncol 2022; 14:17588359221139625. [DOI: 10.1177/17588359221139625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 10/31/2022] [Indexed: 11/30/2022] Open
Abstract
Gastroesophageal cancers (GEC) have a poor survival rate of 20–30% at 5 years, often due to delayed presentations. Neoadjuvant chemoradiotherapy (CRT) followed by surgery or peri-operative chemotherapy and surgery are widely used as the standard of care for patients with resectable GEC. Immune checkpoint inhibitors (ICIs) have improved survival in metastatic and recurrent GEC which led to their application in resectable GEC. Based on the pivotal CheckMate 577 study results, the Food and Drug Administration (FDA) approved nivolumab for patients with completely resected high-risk esophageal or gastroesophageal junction cancer (GEJC). Several ongoing trials with many ICIs could potentially improve resectable GEC outcomes. This review explores the rationale for using ICIs in resectable GEC and discusses the significance of reported clinical trials. Finally, we will examine some ongoing clinical trials and the challenges as well as prospects of ICIs in resectable GEC.
Collapse
Affiliation(s)
- Sarbajit Mukherjee
- Department of Medicine – GI Medical Oncology, Roswell Park Comprehensive Cancer Center, Scott Bieler Clinical Science Center, 9th Floor P-934, Elm & Carlton Streets, Buffalo, NY 14263, USA
| | - Kanak Parmar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Elizabeth Smyth
- Department of Oncology, Cambridge University Hospitals National Health Service Foundation Trust, Cambridge, UK
| |
Collapse
|
39
|
Zhou C, Cheng X, Tu S. Current status and future perspective of immune checkpoint inhibitors in colorectal cancer. Cancer Lett 2021; 521:119-129. [PMID: 34464671 DOI: 10.1016/j.canlet.2021.07.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/23/2021] [Accepted: 07/15/2021] [Indexed: 12/16/2022]
Abstract
Immune checkpoint inhibitors (ICIs), as a subverter of immunotherapy in oncology, are changing all aspects of therapy for malignant tumors, especially their remarkable effects on melanoma and non-small cell lung cancer (NSCLC). For colorectal cancer (CRC), only a small number of patients with high immunogenicity (microsatellite instability-high/mismatch-repair deficient (MSI-H/dMMR)) benefit greatly from ICIs treatment, and most CRC patients with low immunogenicity (microsatellite instability-low/mismatch-repair proficient (MSI-L/pMMR)) do not. Currently, numerous clinical trials are ongoing to improve CRC patients' response to ICIs immunotherapy through better patient selection and novel combination strategies. Thus, this review discusses the current status and latest progress of ICIs treatment in CRC. We expect that these studies can change the pattern of CRC immunotherapy in the future.
Collapse
Affiliation(s)
- Cong Zhou
- Department of Oncology, Renji Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Xiaojiao Cheng
- Department of Oncology, Renji Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China; State Key Laboratory of Oncogenesis and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shuiping Tu
- Department of Oncology, Renji Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China; State Key Laboratory of Oncogenesis and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
40
|
Hofman P. EGFR Status Assessment for Better Care of Early Stage Non-Small Cell Lung Carcinoma: What Is Changing in the Daily Practice of Pathologists? Cells 2021; 10:2157. [PMID: 34440926 PMCID: PMC8392580 DOI: 10.3390/cells10082157] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 02/08/2023] Open
Abstract
The recent emergence of novel neoadjuvant and/or adjuvant therapies for early stage (I-IIIA) non-small cell lung carcinoma (NSCLC), mainly tyrosine kinase inhibitors (TKIs) targeting EGFR mutations and immunotherapy or chemo-immunotherapy, has suddenly required the evaluation of biomarkers predictive of the efficacy of different treatments in these patients. Currently, the choice of one or another of these treatments mainly depends on the results of immunohistochemistry for PD-L1 and of the status of EGFR and ALK. This new development has led to the setup of different analyses for clinical and molecular pathology laboratories, which have had to rapidly integrate a number of new challenges into daily practice and to establish new organization for decision making. This review outlines the impact of the management of biological samples in laboratories and discusses perspectives for pathologists within the framework of EGFR TKIs in early stage NSCLC.
Collapse
Affiliation(s)
- Paul Hofman
- Laboratory of Clinical and Experimental Pathology, CHU Nice, FHU OncoAge, Pasteur Hospital, Université Côte d’Azur, 06108 Nice, France; ; Tel.: +33-492-038-855; Fax: +33-492-8850
- CHU Nice, FHU OncoAge, Hospital-Integrated Biobank BB-0033-00025, Université Côte d’Azur, 06000 Nice, France
| |
Collapse
|
41
|
Bilusic M, Gulley JL. Neoadjuvant Immunotherapy: An Evolving Paradigm Shift? J Natl Cancer Inst 2021; 113:799-800. [PMID: 33432346 DOI: 10.1093/jnci/djaa217] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 12/08/2020] [Indexed: 12/26/2022] Open
Affiliation(s)
- Marijo Bilusic
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - James L Gulley
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
42
|
Meireson A, Tavernier SJ, Van Gassen S, Sundahl N, Demeyer A, Spaas M, Kruse V, Ferdinande L, Van Dorpe J, Hennart B, Allorge D, Haerynck F, Decaestecker K, Rottey S, Saeys Y, Ost P, Brochez L. Immune Monitoring in Melanoma and Urothelial Cancer Patients Treated with Anti-PD-1 Immunotherapy and SBRT Discloses Tumor Specific Immune Signatures. Cancers (Basel) 2021; 13:cancers13112630. [PMID: 34071888 PMCID: PMC8198315 DOI: 10.3390/cancers13112630] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/21/2021] [Accepted: 05/25/2021] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Currently available biomarkers for response to checkpoint inhibitors are incomplete and predominantly focus on tumor tissue analysis e.g., tumor mutational burden, programmed cell death-ligand 1 (PD-L1) expression. Biomarkers in peripheral blood would allow a more dynamic monitoring and could offer a way for sequential adaptation of treatment strategy. We conducted an in-depth analysis of baseline and on-treatment systemic immune features in a cohort of stage III/IV melanoma and stage IV urothelial cancer (UC) patients treated with anti-programmed cell death-1 (anti-PD-1) therapy combined with stereotactic body radiotherapy (SBRT) in a similar regimen/schedule. Baseline immunity was clearly different between these two cohorts, indicating a less active immune landscape in UC patients. This study also detected signatures of proliferation in the CD8+ T-cell compartment pre-treatment and early after anti-PD-1 initiation that were positively correlated with clinical outcome in both tumor types. In addition our data support the biological relevance of PD-1/PD-L1 expression on circulating immune cell subsets, especially in melanoma. Abstract (1) Background: Blockade of the PD-1/PD-L1 pathway has revolutionized the oncology field in the last decade. However, the proportion of patients experiencing a durable response is still limited. In the current study, we performed an extensive immune monitoring in patients with stage III/IV melanoma and stage IV UC who received anti-PD-1 immunotherapy with SBRT. (2) Methods: In total 145 blood samples from 38 patients, collected at fixed time points before and during treatment, were phenotyped via high-parameter flow cytometry, luminex assay and UPLC-MS/MS. (3) Results: Baseline systemic immunity in melanoma and UC patients was different with a more prominent myeloid compartment and a higher neutrophil to lymphocyte ratio in UC. Proliferation (Ki67+) of CD8+ T-cells and of the PD-1+/PD-L1+ CD8+ subset at baseline correlated with progression free survival in melanoma. In contrast a higher frequency of PD-1/PD-L1 expressing non-proliferating (Ki67−) CD8+ and CD4+ T-cells before treatment was associated with worse outcome in melanoma. In UC, the expansion of Ki67+ CD8+ T-cells and of the PD-L1+ subset relative to tumor burden correlated with clinical outcome. (4) Conclusion: This study reveals a clearly different immune landscape in melanoma and UC at baseline, which may impact immunotherapy response. Signatures of proliferation in the CD8+ T-cell compartment prior to and early after anti-PD-1 initiation were positively correlated with clinical outcome in both cohorts. PD-1/PD-L1 expression on circulating immune cell subsets seems of clinical relevance in the melanoma cohort.
Collapse
Affiliation(s)
- Annabel Meireson
- Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium; (A.M.); (N.S.); (A.D.); (M.S.); (V.K.); (J.V.D.); (K.D.); (S.R.); (Y.S.); (P.O.)
- Dermatology Research Unit, Ghent University Hospital, 9000 Ghent, Belgium
| | - Simon J. Tavernier
- Centre for Primary Immunodeficiency Ghent, Primary Immune Deficiency Research Lab, Department of Internal Medicine and Pediatrics, Jeffrey Modell Diagnosis and Research Centre, Ghent University Hospital, 9000 Ghent, Belgium; (S.J.T.); (F.H.)
- VIB Center for Inflammation Research, Unit of Molecular Signal Transduction in Inflammation, 9000 Ghent, Belgium
| | - Sofie Van Gassen
- VIB Center for Inflammation Research, Unit of Data Mining and Modeling for Biomedicine, 9000 Ghent, Belgium;
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, 9000 Ghent, Belgium
| | - Nora Sundahl
- Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium; (A.M.); (N.S.); (A.D.); (M.S.); (V.K.); (J.V.D.); (K.D.); (S.R.); (Y.S.); (P.O.)
- Department of Radiation Oncology and Experimental Cancer Research, Ghent University Hospital, 9000 Ghent, Belgium
| | - Annelies Demeyer
- Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium; (A.M.); (N.S.); (A.D.); (M.S.); (V.K.); (J.V.D.); (K.D.); (S.R.); (Y.S.); (P.O.)
- Dermatology Research Unit, Ghent University Hospital, 9000 Ghent, Belgium
| | - Mathieu Spaas
- Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium; (A.M.); (N.S.); (A.D.); (M.S.); (V.K.); (J.V.D.); (K.D.); (S.R.); (Y.S.); (P.O.)
- Department of Radiation Oncology and Experimental Cancer Research, Ghent University Hospital, 9000 Ghent, Belgium
| | - Vibeke Kruse
- Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium; (A.M.); (N.S.); (A.D.); (M.S.); (V.K.); (J.V.D.); (K.D.); (S.R.); (Y.S.); (P.O.)
- Department of Medical Oncology, Ghent University Hospital, 9000 Ghent, Belgium
| | | | - Jo Van Dorpe
- Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium; (A.M.); (N.S.); (A.D.); (M.S.); (V.K.); (J.V.D.); (K.D.); (S.R.); (Y.S.); (P.O.)
- Department of Pathology, Ghent University Hospital, 9000 Ghent, Belgium;
| | - Benjamin Hennart
- Unité Fonctionnelle de Toxicologie, CHU Lille, F-59000 Lille, France; (B.H.); (D.A.)
- ULR 4483-IMPact de l’Environnement Chimique sur la Santé Humaine (IMPECS), Université de Lille, F-59000 Lille, France
| | - Delphine Allorge
- Unité Fonctionnelle de Toxicologie, CHU Lille, F-59000 Lille, France; (B.H.); (D.A.)
- ULR 4483-IMPact de l’Environnement Chimique sur la Santé Humaine (IMPECS), Université de Lille, F-59000 Lille, France
| | - Filomeen Haerynck
- Centre for Primary Immunodeficiency Ghent, Primary Immune Deficiency Research Lab, Department of Internal Medicine and Pediatrics, Jeffrey Modell Diagnosis and Research Centre, Ghent University Hospital, 9000 Ghent, Belgium; (S.J.T.); (F.H.)
| | - Karel Decaestecker
- Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium; (A.M.); (N.S.); (A.D.); (M.S.); (V.K.); (J.V.D.); (K.D.); (S.R.); (Y.S.); (P.O.)
- Department of Urology, Ghent University Hospital, 9000 Ghent, Belgium
| | - Sylvie Rottey
- Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium; (A.M.); (N.S.); (A.D.); (M.S.); (V.K.); (J.V.D.); (K.D.); (S.R.); (Y.S.); (P.O.)
- Department of Medical Oncology, Ghent University Hospital, 9000 Ghent, Belgium
| | - Yvan Saeys
- Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium; (A.M.); (N.S.); (A.D.); (M.S.); (V.K.); (J.V.D.); (K.D.); (S.R.); (Y.S.); (P.O.)
- VIB Center for Inflammation Research, Unit of Data Mining and Modeling for Biomedicine, 9000 Ghent, Belgium;
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, 9000 Ghent, Belgium
| | - Piet Ost
- Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium; (A.M.); (N.S.); (A.D.); (M.S.); (V.K.); (J.V.D.); (K.D.); (S.R.); (Y.S.); (P.O.)
- Department of Radiation Oncology and Experimental Cancer Research, Ghent University Hospital, 9000 Ghent, Belgium
| | - Lieve Brochez
- Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium; (A.M.); (N.S.); (A.D.); (M.S.); (V.K.); (J.V.D.); (K.D.); (S.R.); (Y.S.); (P.O.)
- Dermatology Research Unit, Ghent University Hospital, 9000 Ghent, Belgium
- Correspondence:
| |
Collapse
|