1
|
Anvari S, Nikbakht M, Vaezi M, Amini-Kafiabad S, Ahmadvand M. Immune checkpoints and ncRNAs: pioneering immunotherapy approaches for hematological malignancies. Cancer Cell Int 2024; 24:410. [PMID: 39702293 DOI: 10.1186/s12935-024-03596-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 12/03/2024] [Indexed: 12/21/2024] Open
Abstract
Hematological malignancies are typically treated with chemotherapy and radiotherapy as the first-line conventional therapies. However, non-coding RNAs (ncRNAs) are a rapidly expanding field of study in cancer biology that influences the growth, differentiation, and proliferation of tumors by targeting immunological checkpoints. This study reviews the results of studies (from 2012 to 2024) that consider the immune checkpoints and ncRNAs in relation to hematological malignancies receiving immunotherapy. This article provides a summary of the latest advancements in immunotherapy for treating hematological malignancies, focusing on the role of immune checkpoints and ncRNAs in the immune response and their capacity for innovative strategies. The paper also discusses the function of immune checkpoints in maintaining immune homeostasis and how their dysregulation can contribute to developing leukemia and lymphoma. Finally, this research concludes with a discussion on the obstacles and future directions in this rapidly evolving field, emphasizing the need for continued research to fully harness the capacity of immune checkpoints and ncRNAs in immunotherapy for hematological malignancies.
Collapse
Affiliation(s)
- Samira Anvari
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Mohsen Nikbakht
- Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Vaezi
- Hematology, Oncology, and Stem Cell Transplantation Research Center Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Sedigheh Amini-Kafiabad
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran.
| | - Mohammad Ahmadvand
- Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
El-Tanani M, Rabbani SA, Ali AA, Alfaouri IGA, Al Nsairat H, Al-Ani IH, Aljabali AA, Rizzo M, Patoulias D, Khan MA, Parvez S, El-Tanani Y. Circadian rhythms and cancer: implications for timing in therapy. Discov Oncol 2024; 15:767. [PMID: 39692981 DOI: 10.1007/s12672-024-01643-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 11/27/2024] [Indexed: 12/19/2024] Open
Abstract
Circadian rhythms, intrinsic cycles spanning approximately 24 h, regulate numerous physiological processes, including sleep-wake cycles, hormone release, and metabolism. These rhythms are orchestrated by the circadian clock, primarily located in the suprachiasmatic nucleus (SCN) of the hypothalamus. Disruptions in circadian rhythms, whether due to genetic mutations, environmental factors, or lifestyle choices, can significantly impact health, contributing to disorders such as sleep disturbances, metabolic syndrome, and cardiovascular diseases. Additionally, there is a profound link between the disruption of circadian rhythms and development of various cancer, the influence on disease incidence and progression. This incurred regulation by circadian clock on pathways has its implication in tumorigenesis, such as cell cycle control, DNA damage response, apoptosis, and metabolism. Furthermore, the circadian timing system modulates the efficacy and toxicity of cancer treatments. In cancer treatment, the use of chronotherapy to optimize the timing of medical treatments, involves administering chemotherapy, radiation, or other therapeutic interventions at specific intervals to enhance efficacy and minimize side effects. This approach capitalizes on the circadian variations in cellular processes, including DNA repair, cell cycle progression, and drug metabolism. Preclinical and clinical studies have demonstrated that chronotherapy can significantly improve the therapeutic index of chemotherapeutic agents like cisplatin and 5-fluorouracil by enhancing anticancer activity and reducing toxicity. Further research is needed to elucidate the mechanisms underlying circadian regulation of cancer and to develop robust chronotherapeutic protocols tailored to individual patients' circadian profiles, potentially transforming cancer care into more effective and personalized treatment strategies.
Collapse
Affiliation(s)
- Mohamed El-Tanani
- RAK College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates.
- Translational and Medical Research Centre (TMRC), Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates.
| | - Syed Arman Rabbani
- RAK College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
- Translational and Medical Research Centre (TMRC), Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | - Areeg Anwer Ali
- RAK College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
- Translational and Medical Research Centre (TMRC), Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | - Ibrahim Ghaleb Ali Alfaouri
- Translational and Medical Research Centre (TMRC), Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
- RAK College of Nursing, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | - Hamdi Al Nsairat
- Pharmacological and Diagnostic Research Center, Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Israa Hamid Al-Ani
- Pharmacological and Diagnostic Research Center, Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Alaa A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Pharmacy, Yarmouk University, Irbid, Jordan
| | - Manfredi Rizzo
- Department of Health Promotion, Mother and Childcare, Internal Medicine and Medical Specialties, School of Medicine, University of Palermo, Palermo, Italy
| | - Dimitrios Patoulias
- Second Department of Cardiology, Aristotle University of Thessaloniki, Hippokration General Hospital, Athens, Greece
- Outpatient Department of Cardiometabolic Medicine, Second Department of Cardiology, Aristotle University of Thessaloniki, Hippokration General Hospital, Athens, Greece
| | - Mohammad Ahmed Khan
- School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Suhel Parvez
- School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | | |
Collapse
|
3
|
Berardi R, Chiariotti R, Mentrasti G. Is nutritional status a new indicator to use in clinical practice for colorectal cancer patients? World J Gastrointest Oncol 2024; 16:4537-4542. [PMID: 39678789 PMCID: PMC11577381 DOI: 10.4251/wjgo.v16.i12.4537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/20/2024] [Accepted: 07/15/2024] [Indexed: 11/12/2024] Open
Abstract
In this editorial we comment on the interesting article by Liu et al. The topic of discussion is the need for a cost-effective and easy-to-use scoring system for predicting the prognosis of colorectal cancer patients. In this context, nutritional assessment plays a crucial role in the multimodal evaluation of patients. In particular, the controlling nutritional status score was found to be an effective tool in the clinical decision-making process, in order to customize treatment strategies and to improve patient outcomes.
Collapse
Affiliation(s)
- Rossana Berardi
- Department of Medical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria delle Marche, Ancona 60126, Marche, Italy
| | - Rebecca Chiariotti
- Department of Medical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria delle Marche, Ancona 60126, Marche, Italy
| | - Giulia Mentrasti
- Department of Medical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria delle Marche, Ancona 60126, Marche, Italy
| |
Collapse
|
4
|
Chen X, Ding W, Jiang Y, Shi W, Qiu Y, Zhao H, Luo X. Emerging Strategies for Local Delivery of Immune Checkpoint Inhibitors to Potentiate Cancer Immunotherapy: Current Status and Future Prospects. ACS APPLIED MATERIALS & INTERFACES 2024; 16:59682-59696. [PMID: 39436983 DOI: 10.1021/acsami.4c12603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Cancer constitutes a significant threat to patients' lives worldwide. Immunotherapy, particularly immune checkpoint inhibitors (ICIs) that boost antitumor immunity by targeting immune checkpoint components, has emerged as a promising strategy for its treatment in recent years. However, the objective response rates of the ICIs are unsatisfactory. As the primary route, systemic administration of ICIs is often accompanied by immune-related adverse events. Local delivery of ICIs serves as a potential therapeutic strategy that can improve the efficacy while simultaneously reducing side effects through precise drug release at the tumor site. Initial validation of direct local application of ICIs for tumors in clinical trials has indicated reduced side effects and improved efficacy, while low bioavailability remains a challenge. Furthermore, research on various carriers, including nanoparticles, microneedles, hydrogels, combined platforms, and implantable devices for local release of ICIs has exhibited applying potential in treating murine tumors, among which combined platforms such as combined hydrogel system hold the highest promise due to their encompassment of the advantages of multiple carriers. These carriers, by incorporating ICIs and other therapeutics, could manage cancers more potently, which needs to be confirmed in clinical trials after the refinement of their biocompatibility. This review summarizes the latest research advancements regarding local administration of ICIs, with a particular focus on the carriers for local delivery as well as the combination therapies, thus providing novel insights and research guidance for scholars to enhance the efficacy of locally delivered ICIs on managing multiple cancers in the future.
Collapse
Affiliation(s)
- Xin Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Wei Ding
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yuchen Jiang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Wenjin Shi
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yan Qiu
- Department of Pathology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hang Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xiaobo Luo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
5
|
Moon S, Jung M, Go S, Hong J, Sohn HS, Kim C, Kang M, Lee BJ, Kim J, Lim J, Kim BS. Engineered Nanoparticles for Enhanced Antitumoral Synergy Between Macrophages and T Cells in the Tumor Microenvironment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2410340. [PMID: 39252658 DOI: 10.1002/adma.202410340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/22/2024] [Indexed: 09/11/2024]
Abstract
T cells and macrophages have the potential to collaborate to eliminate tumor cells efficiently. Macrophages can eliminate tumor cells through phagocytosis and subsequently activate T cells by presenting tumor antigens. The activated T cells, in turn, can kill tumor cells and redirect tumor-associated macrophages toward an antitumoral M1 phenotype. However, checkpoint molecules expressed on tumor cells impede the collaborative action of these immune cells. Meanwhile, monotherapy with a single immune checkpoint inhibitor (ICI) for either macrophages or T cells yields suboptimal efficacy in cancer patients. To address this challenge, here a nanoparticle capable of efficiently delivering dual ICIs to tumors for both macrophages and T cells is developed. These programmed cell death protein 1 (PD-1)-transfected macrophage membrane-derived nanoparticles (PMMNPs) can target tumors and provide signal-regulatory protein alpha and PD-1 to block CD47 and programmed cell death-ligand 1 (PD-L1), respectively, on tumor cells. PMMNPs enhance macrophage-mediated cancer cell phagocytosis and antigen presentation, promote T cell activation, and induce the reprogramming of macrophages toward an antitumoral phenotype. In syngeneic tumor-bearing mice, PMMNPs demonstrate superior therapeutic efficacy compared to nanoparticles delivering single ICIs and non-targeted delivery of anti-CD47 and anti-PD-L1 antibodies. PMMNPs capable of augmenting the antitumoral interplay between macrophages and T cells may offer a promising avenue for cancer immunotherapy.
Collapse
Affiliation(s)
- Sangjun Moon
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Mungyo Jung
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seokhyeong Go
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jihye Hong
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hee Su Sohn
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Cheesue Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Mikyung Kang
- School of Health and Environmental Science, Korea University, Seoul, 02841, Republic of Korea
| | - Byung Joon Lee
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jungwoo Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jinwoong Lim
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Byung-Soo Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
- Institute of Engineering Research, Institute of Chemical Processes, and BioMAX, Seoul National University, Seoul, 08826, Republic of Korea
| |
Collapse
|
6
|
Gamero MT, Patel A, Storozynsky E. The Good (Tumor Killing) and the Bad (Cardiovascular Complications) of Immunologic Checkpoint Inhibitors. Curr Cardiol Rep 2024:10.1007/s11886-024-02147-x. [PMID: 39441327 DOI: 10.1007/s11886-024-02147-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/23/2024] [Indexed: 10/25/2024]
Abstract
PURPOSE OF REVIEW This review details the significant advancement in knowledge of Immune-checkpoint inhibitor (ICI) and its potential deleterious cardiac immune-related adverse effects (irAE). We explore their mechanisms on the cardiac tissue, providing guidance on risk factors, clinical presentations, diagnostic strategies along with treatment. RECENT FINDINGS Recent findings have provided insights of cardiac irAEs that exist beyond the previously well-known ICI-induced myocarditis. We have a better understanding of the wide variety of cardiac irAEs pathologies both early and late onset. Moreover, there is more data on mechanisms of cardiotoxicity and patient and therapy-related risk factors, supporting closer routine cardiac monitoring with biomarkers and imaging for prevention and early detection. Diagnosing cardiac irAEs is a challenge given its broad clinical presentation. A high-level of suspicion in addition to early work-up is crucial to prevent serious cardiac events. A multi-disciplinary team including Cardiologists and Oncologists is essential for closely monitor patients' cardiac status on ICI therapy. There is a need of updated guidelines to establish clear recommendations in patients on ICIs.
Collapse
Affiliation(s)
- Maria T Gamero
- Department of Medicine, Division of Cardiovascular Disease, Jefferson Heart Institute, Thomas Jefferson University Hospital, Philadelphia, PA, USA.
| | - Avish Patel
- Department of Medicine, Division of Cardiovascular Disease, Jefferson Heart Institute, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - Eugene Storozynsky
- Department of Medicine, Division of Cardiovascular Disease, Jefferson Heart Institute, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| |
Collapse
|
7
|
Wang Z, Sun Z, Lv H, Wu W, Li H, Jiang T. Machine learning-based model for CD4 + conventional T cell genes to predict survival and immune responses in colorectal cancer. Sci Rep 2024; 14:24426. [PMID: 39424871 PMCID: PMC11489786 DOI: 10.1038/s41598-024-75270-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 10/03/2024] [Indexed: 10/21/2024] Open
Abstract
Globally, CRC ranks as a principal cause of mortality, with projections indicating a substantial rise in both incidence and mortality by the year 2040. The immunological responses to cancer heavily rely on the function of CD4Tconv. Despite this critical role, prognostic studies on CRC-related CD4Tconv remain insufficient. In this investigation, transcriptomic and clinical data were sourced from TCGA and GEO. Initially, we pinpointed CD4TGs using single-cell datasets. Prognostic genes were then isolated through univariate Cox regression analysis. Building upon this, 101 machine learning algorithms were employed to devise a novel risk assessment framework, which underwent rigorous validation using Kaplan-Meier survival analysis, univariate and multivariate Cox regression, time-dependent ROC curves, nomograms, and calibration plots. Furthermore, GSEA facilitated the examination of these genes' potential roles. The RS derived from this model was also analyzed for its implications in the TME, and its potential utility in immunotherapy and chemotherapy contexts. A novel prognostic model was developed, utilizing eight CD4TGs that are significantly linked to the outcomes of patients with CRC. This model's RS showcased remarkable predictive reliability for the overall survival rates of CRC patients and strongly correlated with malignancy levels. RS serves as an autonomous prognostic indicator, capable of accurately forecasting patient prognoses. Based on the median value of RS, patients were categorized into subgroups of high and low risk. The subgroup with higher risk demonstrated increased immune infiltration and heightened activity of genes associated with immunity. This investigation's establishment of a CD4TGs risk model introduces novel biomarkers for the clinical evaluation of CRC risks. These biomarkers may enhance therapeutic approaches and, in turn, elevate the clinical outcomes for patients with CRC by facilitating an integrated treatment strategy.
Collapse
Affiliation(s)
- Zijing Wang
- First Clinical Medical College, General Hospital of Ningxia Medical University, 804 Shengli Road, Yinchuan, 750004, China
| | - Zhanyuan Sun
- First Clinical Medical College, General Hospital of Ningxia Medical University, 804 Shengli Road, Yinchuan, 750004, China
| | - Hengyi Lv
- First Clinical Medical College, General Hospital of Ningxia Medical University, 804 Shengli Road, Yinchuan, 750004, China
| | - Wenjun Wu
- First Clinical Medical College, General Hospital of Ningxia Medical University, 804 Shengli Road, Yinchuan, 750004, China
| | - Hai Li
- Department of Anal-Colorectal Surgery, General Hospital of Ningxia Medical University, 804 Shengli Road, Yinchuan, 750004, China
| | - Tao Jiang
- Department of Anal-Colorectal Surgery, General Hospital of Ningxia Medical University, 804 Shengli Road, Yinchuan, 750004, China.
| |
Collapse
|
8
|
Iijima H, Sakai A, Ebisumoto K, Ogura G, Yamauchi M, Teramura T, Yamazaki A, Watanabe T, Inagi T, Yanagiya R, Yamamoto A, Ashida H, Ota Y, Sato Y, Kobayashi N, Maki D, Nakamura N, Okami K. Combined Positive Score and Cisplatin Sensitivity Are Prognostic Factors for Response to Nivolumab Therapy for Recurrent Metastatic Squamous Cell Carcinoma of the Head and Neck. Clin Med Insights Oncol 2024; 18:11795549241290030. [PMID: 39429682 PMCID: PMC11490953 DOI: 10.1177/11795549241290030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 09/03/2024] [Indexed: 10/22/2024] Open
Abstract
Background Recurrent or metastatic squamous cell carcinoma of the head and neck (R/MHNSCC) is a challenging malignancy with a poor prognosis and limited treatment options. Nivolumab, an immune checkpoint inhibitor (ICI) targeting the programmed cell death/programmed cell death ligand 1 (PD-1/PD-L1) pathway, has emerged as a promising therapy for these patients. However, identifying biomarkers predictive of response to nivolumab remains critical for optimizing treatment strategies. Previous studies have suggested that PD-L1 expression, as determined by the Combined Positive Score (CPS) and other clinical factors, may influence treatment outcome. This study aims to retrospectively examine whether CPS can be a biomarker by staining PD-L1 with 22 C3 antibody in R/MHNSCC patients treated with nivolumab. Methods This retrospective study reviewed the medical records of R/MHNSCC patients treated with ICIs at Tokai University Hospital from April 2017 to December 2022. We examined the relationship between response rate to ICI therapy, PD-L1 staining, biomarkers, and survival. Statistical analyses included t-test, chi-square test, and Cox regression. Results This study included 92 nivolumab-treated patients. Combined Positive Score was evaluable in 53 of these patients. Patients with a CPS of 15 or higher had better progression-free survival (PFS) (P = .0171), with a median PFS) of 13 months. In the Various Definitions analysis, cisplatin-sensitive patients also had good PFS (P = .0295). The cisplatin-sensitive patient population with a CPS of 15 or higher had the best PFS, with a median of 14 months (P = .006). There was no significant difference in overall survival (OS) by CPS value. Immune-related adverse events did not affect OS or PFS. Conclusions CPS ⩾ 15 and cisplatin sensitivity are promising prognostic markers for nivolumab therapy in R/MHNSCC. Considering these biomarkers in patient selection could maximize the therapeutic benefits of nivolumab. This finding may help to optimize ICI therapy strategies.
Collapse
Affiliation(s)
- Hiroaki Iijima
- Department of Otolaryngology, Head and Neck Surgery, Tokai University School of Medicine, Isehara, Japan
| | - Akihiro Sakai
- Department of Otolaryngology, Head and Neck Surgery, Tokai University School of Medicine, Isehara, Japan
| | - Koji Ebisumoto
- Department of Otolaryngology, Head and Neck Surgery, Tokai University School of Medicine, Isehara, Japan
| | - Go Ogura
- Department of Pathology, Tokai University School of Medicine, Isehara, Japan
| | - Mayu Yamauchi
- Department of Otolaryngology, Head and Neck Surgery, Tokai University School of Medicine, Isehara, Japan
| | - Takanobu Teramura
- Department of Otolaryngology, Head and Neck Surgery, Tokai University School of Medicine, Isehara, Japan
| | - Aritomo Yamazaki
- Department of Otolaryngology, Head and Neck Surgery, Tokai University School of Medicine, Isehara, Japan
| | - Takane Watanabe
- Department of Otolaryngology, Head and Neck Surgery, Tokai University School of Medicine, Isehara, Japan
| | - Toshihide Inagi
- Department of Otolaryngology, Head and Neck Surgery, Tokai University School of Medicine, Isehara, Japan
| | - Ryoko Yanagiya
- Department of Otolaryngology, Head and Neck Surgery, Tokai University School of Medicine, Isehara, Japan
| | - Ai Yamamoto
- Department of Otolaryngology, Head and Neck Surgery, Tokai University School of Medicine, Isehara, Japan
| | - Hiroshi Ashida
- Department of Otolaryngology, Head and Neck Surgery, Tokai University School of Medicine, Isehara, Japan
| | - Yoshiyuki Ota
- Department of Otolaryngology, Head and Neck Surgery, Tokai University School of Medicine, Isehara, Japan
| | - Yurina Sato
- Department of Otolaryngology, Head and Neck Surgery, Tokai University School of Medicine, Isehara, Japan
| | - Naoya Kobayashi
- Department of Otolaryngology, Head and Neck Surgery, Tokai University School of Medicine, Isehara, Japan
| | - Daisuke Maki
- Department of Otolaryngology, Head and Neck Surgery, Tokai University School of Medicine, Isehara, Japan
| | - Naoya Nakamura
- Department of Pathology, Tokai University School of Medicine, Isehara, Japan
| | - Kenji Okami
- Department of Otolaryngology, Head and Neck Surgery, Tokai University School of Medicine, Isehara, Japan
| |
Collapse
|
9
|
Medina A, Carballo J, González‐Marcano E, Blanca I, Convit AF. Breast cancer immunotherapy: Realities and advances. CANCER INNOVATION 2024; 3:e140. [PMID: 39308754 PMCID: PMC11416644 DOI: 10.1002/cai2.140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 05/09/2024] [Accepted: 05/21/2024] [Indexed: 09/25/2024]
Abstract
Breast cancer (BC) is the most common malignant tumor and the main cause of death in women worldwide. With increased knowledge regarding tumor escape mechanisms and advances in immunology, many new antitumor strategies such as nonspecific immunotherapies, monoclonal antibodies, anticancer vaccines, and oncolytic viruses, among others, make immunotherapy a promising approach for the treatment of BC. However, these approaches still require meticulous assessment and readjustment as resistance and modest response rates remain important barriers. In this article, we aim to summarize the most recent data available in BC immunotherapy to include the results of ongoing clinical trials and approved therapies used as monotherapies or in combination with conventional treatments.
Collapse
Affiliation(s)
- Aixa Medina
- Jacinto Convit World Organization Inc.Pompano BeachFloridaUSA
- Facultad de MedicinaUniversidad Central de VenezuelaCaracasVenezuela
| | | | | | - Isaac Blanca
- Unidad Experimental de InmunoterapiaFundación Jacinto ConvitCaracasVenezuela
| | - Ana F. Convit
- Jacinto Convit World Organization Inc.Pompano BeachFloridaUSA
- Unidad Experimental de InmunoterapiaFundación Jacinto ConvitCaracasVenezuela
| |
Collapse
|
10
|
Mao W, Yoo HS. Inorganic Nanoparticle Functionalization Strategies in Immunotherapeutic Applications. Biomater Res 2024; 28:0086. [PMID: 39323561 PMCID: PMC11423863 DOI: 10.34133/bmr.0086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/20/2024] [Accepted: 09/05/2024] [Indexed: 09/27/2024] Open
Abstract
Nanotechnology has been increasingly utilized in anticancer treatment owing to its ability of engineering functional nanocarriers that enhance therapeutic effectiveness while minimizing adverse effects. Inorganic nanoparticles (INPs) are prevalent nanocarriers to be customized for a wide range of anticancer applications, including theranostics, imaging, targeted drug delivery, and therapeutics, because they are advantageous for their superior biocompatibility, unique optical properties, and capacity of being modified via versatile surface functionalization strategies. In the past decades, the high adaptation of INPs in this emerging immunotherapeutic field makes them good carrier options for tumor immunotherapy and combination immunotherapy. Tumor immunotherapy requires targeted delivery of immunomodulating therapeutics to tumor locations or immunological organs to provoke immune cells and induce tumor-specific immune response while regulating immune homeostasis, particularly switching the tumor immunosuppressive microenvironment. This review explores various INP designs and formulations, and their employment in tumor immunotherapy and combination immunotherapy. We also introduce detailed demonstrations of utilizing surface engineering tactics to create multifunctional INPs. The generated INPs demonstrate the abilities of stimulating and enhancing the immune response, specific targeting, and regulating cancer cells, immune cells, and their resident microenvironment, sometimes along with imaging and tracking capabilities, implying their potential in multitasking immunotherapy. Furthermore, we discuss the promises of INP-based combination immunotherapy in tumor treatments.
Collapse
Affiliation(s)
- Wei Mao
- Department of Biomedical Materials Engineering, Kangwon National University, Chuncheon 24341, Republic of Korea
- Institute for Molecular Science and Fusion Technology, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Hyuk Sang Yoo
- Department of Biomedical Materials Engineering, Kangwon National University, Chuncheon 24341, Republic of Korea
- Institute for Molecular Science and Fusion Technology, Kangwon National University, Chuncheon 24341, Republic of Korea
- Institute of Biomedical Science, Kangwon National University, Chuncheon 24341, Republic of Korea
- Kangwon Radiation Convergence Research Center, Kangwon National University, Chuncheon 24341, Republic of Korea
| |
Collapse
|
11
|
Farahani H, Darvishvand R, Khademolhosseini A, Erfani N. Unwrapping the immunological alterations in testicular germ cell tumors: From immune homeostasis to malignancy and emerging immunotherapies. Andrology 2024. [PMID: 39253799 DOI: 10.1111/andr.13751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 08/18/2024] [Accepted: 08/20/2024] [Indexed: 09/11/2024]
Abstract
BACKGROUND Testicular germ cell tumors (TGCTs), derived from primordial germ cells, are rare malignancies with high curative potential. However, the emergence of new evidence indicating that 15% of patients experience tumor progression, leading to death, underscores the need for innovative therapeutics. OBJECTIVES This review aimed to explore the immune status in maintaining testicular health and the immune-related aspects of malignancy. Furthermore, it presents an overview of current data on the use of immunotherapy for TGCT patients. RESULTS DISCUSSION Recent advances in immunology have opened a promising avenue for studying diseases and highlighted its role in treating diseases. While the immunopathological facets of TGCTs are not fully understood, investigations suggest a complex interplay among testis-resident immune cells, testis-specific cells (i.e., Sertoli cells (SCs) and Leydig cells (LCs)), and immune-regulating mediators (e.g., sex hormones) in the normal testicle that foster the testicular immune privilege (TIP). Although TIP plays a crucial role in sperm production, it also makes testis vulnerable to tumor development. In the context of cancer-related inflammation, disruption of TIP leads to an imbalanced immune response, resulting in chronic inflammation that can contribute to testicular tissue dysfunction or loss, potentially aiding in cancer invasion and progression. CONCLUSION Comparing the immune profiles of normal and malignant testes is valuable and may provide insights into different aspects of testicular immunity and immune-based treatment approaches. For patients resistant to chemotherapy and with a poor prognosis, immunotherapy has shown promising results. However, its effectiveness in treating resistant TGCTs or preventing tumor recurrence is still uncertain.
Collapse
Affiliation(s)
- Hadiseh Farahani
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Darvishvand
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Aida Khademolhosseini
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nasrollah Erfani
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
12
|
Wang YY, Song JJ. A case report of the diagnosis and treatment of immune checkpoint inhibitor-related encephalitis induced by camrelizumab. AME Case Rep 2024; 8:101. [PMID: 39380870 PMCID: PMC11459425 DOI: 10.21037/acr-24-58] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 07/26/2024] [Indexed: 10/10/2024]
Abstract
Background Camrelizumab has been widely used in the treatment of various cancers, it is important to determine the side-effect of this drug and the corresponding treatment strategy. Case Description The current case report describes the clinic, diagnosis, treatment and prognosis of camrelizumab-related encephalitis. Camrelizumab was administrated to a 67-year-old man with squamous cell carcinoma (SCC), a form of non-small cell lung cancer (NSCLC). One month after the treatment, the patient showed typical encephalitis symptoms including systemic fatigue, numbness of extremities and walking instability. Furthermore, the total protein in cerebrospinal fluid (CSF) was significantly elevated (1,399 vs. normal range 120-600 mg/L). Importantly, magnetic resonance imaging showed there was no brain metastasis. The patient did not get better after two days of intravenous injection of thioctic acid (1.2 g) and cobamamide (1.5 mg) once daily. Therefore, this patient was diagnosed as camrelizumab-related encephalitis. Then, we put him on one-month regimen: oral taper corticoids (methylprednisolone, MP) at 500 mg (days 1-4), 120 mg (days 5-10) and 60 mg (days 11-15); MP was replaced with oral prednisone acetate at 30 mg (days 16-30). After the treatment, the total protein in CSF was decreased to 873 mg/L, and all of encephalitis-related symptom was completely lost. About one year after the onset of encephalitis, the patient showed no recurrence of neurological symptoms. Conclusions The present case proves the efficacy and safety of corticoids in the treatment of camrelizumab-related adverse effects.
Collapse
Affiliation(s)
- Ya-Yun Wang
- Department of Pharmacy, The First People’s Hospital of Jiashan, Jiaxing, China
| | - Jian-Jiang Song
- Department of Cardiovascular Medicine, The First People’s Hospital of Jiashan, Jiaxing, China
| |
Collapse
|
13
|
Pavelescu LA, Enache RM, Roşu OA, Profir M, Creţoiu SM, Gaspar BS. Predictive Biomarkers and Resistance Mechanisms of Checkpoint Inhibitors in Malignant Solid Tumors. Int J Mol Sci 2024; 25:9659. [PMID: 39273605 PMCID: PMC11395316 DOI: 10.3390/ijms25179659] [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: 08/22/2024] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 09/15/2024] Open
Abstract
Predictive biomarkers for immune checkpoint inhibitors (ICIs) in solid tumors such as melanoma, hepatocellular carcinoma (HCC), colorectal cancer (CRC), non-small cell lung cancer (NSCLC), endometrial carcinoma, renal cell carcinoma (RCC), or urothelial carcinoma (UC) include programmed cell death ligand 1 (PD-L1) expression, tumor mutational burden (TMB), defective deoxyribonucleic acid (DNA) mismatch repair (dMMR), microsatellite instability (MSI), and the tumor microenvironment (TME). Over the past decade, several types of ICIs, including cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) inhibitors, anti-programmed cell death 1 (PD-1) antibodies, anti-programmed cell death ligand 1 (PD-L1) antibodies, and anti-lymphocyte activation gene-3 (LAG-3) antibodies have been studied and approved by the Food and Drug Administration (FDA), with ongoing research on others. Recent studies highlight the critical role of the gut microbiome in influencing a positive therapeutic response to ICIs, emphasizing the importance of modeling factors that can maintain a healthy microbiome. However, resistance mechanisms can emerge, such as increased expression of alternative immune checkpoints, T-cell immunoglobulin (Ig), mucin domain-containing protein 3 (TIM-3), LAG-3, impaired antigen presentation, and alterations in the TME. This review aims to synthesize the data regarding the interactions between microbiota and immunotherapy (IT). Understanding these mechanisms is essential for optimizing ICI therapy and developing effective combination strategies.
Collapse
Affiliation(s)
- Luciana Alexandra Pavelescu
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Robert Mihai Enache
- Department of Radiology and Medical Imaging, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Oana Alexandra Roşu
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Oncology, Elias University Emergency Hospital, 011461 Bucharest, Romania
| | - Monica Profir
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Oncology, Elias University Emergency Hospital, 011461 Bucharest, Romania
| | - Sanda Maria Creţoiu
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Bogdan Severus Gaspar
- Department of Surgery, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Surgery Clinic, Bucharest Emergency Clinical Hospital, 014461 Bucharest, Romania
| |
Collapse
|
14
|
Buehning F, Lerchner T, Vogel J, Hendgen-Cotta UB, Totzeck M, Rassaf T, Michel L. Preclinical models of cardiotoxicity from immune checkpoint inhibitor therapy. Basic Res Cardiol 2024:10.1007/s00395-024-01070-0. [PMID: 39039301 DOI: 10.1007/s00395-024-01070-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 06/30/2024] [Accepted: 07/16/2024] [Indexed: 07/24/2024]
Abstract
Immune checkpoint inhibitor (ICI) therapy represents a ground-breaking paradigm in cancer treatment, harnessing the immune system to combat malignancies by targeting checkpoints such as cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein 1 (PD-1). The use of ICI therapy generates distinctive immune-related adverse events (irAEs) including cardiovascular toxicity, necessitating targeted research efforts. This comprehensive review explores preclinical models dedicated to ICI-mediated cardiovascular complications including myocarditis. Tailored preclinical models of ICI-mediated myocardial toxicities highlight the key role of CD8+ T cells, emphasizing the profound impact of immune checkpoints on maintaining cardiac integrity. Cytokines and macrophages were identified as possible driving factors in disease progression, and at the same time, initial data on possible cardiac antigens responsible are emerging. The implications of contributing factors including thoracic radiation, autoimmune disorder, and the presence of cancer itself are increasingly understood. Besides myocarditis, mouse models unveiled an accelerated progression of atherosclerosis, adding another layer for a thorough understanding of the diverse processes involving cardiovascular immune checkpoint signalling. This review aims to discuss current preclinical models of ICI cardiotoxicity and their potential for improving enhanced risk assessment and diagnostics, offering potential targets for innovative cardioprotective strategies. Lessons from ICI therapy can drive novel approaches in cardiovascular research, extending insights to areas such as myocardial infarction and heart failure.
Collapse
Affiliation(s)
- Florian Buehning
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Tobias Lerchner
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Julia Vogel
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Ulrike B Hendgen-Cotta
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Matthias Totzeck
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Tienush Rassaf
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Lars Michel
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany.
| |
Collapse
|
15
|
Kuznetsova AV, Glukhova XA, Popova OP, Beletsky IP, Ivanov AA. Contemporary Approaches to Immunotherapy of Solid Tumors. Cancers (Basel) 2024; 16:2270. [PMID: 38927974 PMCID: PMC11201544 DOI: 10.3390/cancers16122270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/11/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024] Open
Abstract
In recent years, the arrival of the immunotherapy industry has introduced the possibility of providing transformative, durable, and potentially curative outcomes for various forms of malignancies. However, further research has shown that there are a number of issues that significantly reduce the effectiveness of immunotherapy, especially in solid tumors. First of all, these problems are related to the protective mechanisms of the tumor and its microenvironment. Currently, major efforts are focused on overcoming protective mechanisms by using different adoptive cell therapy variants and modifications of genetically engineered constructs. In addition, a complex workforce is required to develop and implement these treatments. To overcome these significant challenges, innovative strategies and approaches are necessary to engineer more powerful variations of immunotherapy with improved antitumor activity and decreased toxicity. In this review, we discuss recent innovations in immunotherapy aimed at improving clinical efficacy in solid tumors, as well as strategies to overcome the limitations of various immunotherapies.
Collapse
Affiliation(s)
- Alla V. Kuznetsova
- Laboratory of Molecular and Cellular Pathology, Russian University of Medicine (Formerly A.I. Evdokimov Moscow State University of Medicine and Dentistry), Ministry of Health of the Russian Federation, Bld 4, Dolgorukovskaya Str, 1127006 Moscow, Russia; (A.V.K.); (O.P.P.)
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov Street, 119334 Moscow, Russia
| | - Xenia A. Glukhova
- Onni Biotechnologies Ltd., Aalto University Campus, Metallimiehenkuja 10, 02150 Espoo, Finland; (X.A.G.); (I.P.B.)
| | - Olga P. Popova
- Laboratory of Molecular and Cellular Pathology, Russian University of Medicine (Formerly A.I. Evdokimov Moscow State University of Medicine and Dentistry), Ministry of Health of the Russian Federation, Bld 4, Dolgorukovskaya Str, 1127006 Moscow, Russia; (A.V.K.); (O.P.P.)
| | - Igor P. Beletsky
- Onni Biotechnologies Ltd., Aalto University Campus, Metallimiehenkuja 10, 02150 Espoo, Finland; (X.A.G.); (I.P.B.)
| | - Alexey A. Ivanov
- Laboratory of Molecular and Cellular Pathology, Russian University of Medicine (Formerly A.I. Evdokimov Moscow State University of Medicine and Dentistry), Ministry of Health of the Russian Federation, Bld 4, Dolgorukovskaya Str, 1127006 Moscow, Russia; (A.V.K.); (O.P.P.)
| |
Collapse
|
16
|
Arndt P, Turkowski K, Cekay M, Eul B, Grimminger F, Savai R. Endothelin and the tumor microenvironment: a finger in every pie. Clin Sci (Lond) 2024; 138:617-634. [PMID: 38785410 PMCID: PMC11130555 DOI: 10.1042/cs20240426] [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: 03/08/2024] [Revised: 05/07/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024]
Abstract
The tumor microenvironment (TME) plays a central role in the development of cancer. Within this complex milieu, the endothelin (ET) system plays a key role by triggering epithelial-to-mesenchymal transition, causing degradation of the extracellular matrix and modulating hypoxia response, cell proliferation, composition, and activation. These multiple effects of the ET system on cancer progression have prompted numerous preclinical studies targeting the ET system with promising results, leading to considerable optimism for subsequent clinical trials. However, these clinical trials have not lived up to the high expectations; in fact, the clinical trials have failed to demonstrate any substantiated benefit of targeting the ET system in cancer patients. This review discusses the major and recent advances of the ET system with respect to TME and comments on past and ongoing clinical trials of the ET system.
Collapse
Affiliation(s)
- Philipp F. Arndt
- Lung Microenvironmental Niche in Cancerogenesis, Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
- Department of Internal Medicine, Justus-Liebig-University Giessen, Universities of Giessen and Marburg Lung Centre (UGMLC), Member of the Cardio-Pulmonary Institute (CPI), Member of the German Centre for Lung Research (DZL), Giessen, Germany
- Max Planck Institute for Heart and Lung Research, Member of the DZL, Member of the CPI, Bad Nauheim, Germany
| | - Kati Turkowski
- Lung Microenvironmental Niche in Cancerogenesis, Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
- Max Planck Institute for Heart and Lung Research, Member of the DZL, Member of the CPI, Bad Nauheim, Germany
| | - Michael J. Cekay
- Lung Microenvironmental Niche in Cancerogenesis, Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
- Department of Internal Medicine, Justus-Liebig-University Giessen, Universities of Giessen and Marburg Lung Centre (UGMLC), Member of the Cardio-Pulmonary Institute (CPI), Member of the German Centre for Lung Research (DZL), Giessen, Germany
| | - Bastian Eul
- Department of Internal Medicine, Justus-Liebig-University Giessen, Universities of Giessen and Marburg Lung Centre (UGMLC), Member of the Cardio-Pulmonary Institute (CPI), Member of the German Centre for Lung Research (DZL), Giessen, Germany
| | - Friedrich Grimminger
- Lung Microenvironmental Niche in Cancerogenesis, Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
- Department of Internal Medicine, Justus-Liebig-University Giessen, Universities of Giessen and Marburg Lung Centre (UGMLC), Member of the Cardio-Pulmonary Institute (CPI), Member of the German Centre for Lung Research (DZL), Giessen, Germany
| | - Rajkumar Savai
- Lung Microenvironmental Niche in Cancerogenesis, Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
- Department of Internal Medicine, Justus-Liebig-University Giessen, Universities of Giessen and Marburg Lung Centre (UGMLC), Member of the Cardio-Pulmonary Institute (CPI), Member of the German Centre for Lung Research (DZL), Giessen, Germany
- Max Planck Institute for Heart and Lung Research, Member of the DZL, Member of the CPI, Bad Nauheim, Germany
| |
Collapse
|
17
|
Mancarella C, Morrione A, Scotlandi K. Extracellular Interactors of the IGF System: Impact on Cancer Hallmarks and Therapeutic Approaches. Int J Mol Sci 2024; 25:5915. [PMID: 38892104 PMCID: PMC11172729 DOI: 10.3390/ijms25115915] [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: 05/09/2024] [Revised: 05/24/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
Dysregulation of the insulin-like growth factor (IGF) system determines the onset of various pathological conditions, including cancer. Accordingly, therapeutic strategies have been developed to block this system in tumor cells, but the results of clinical trials have been disappointing. After decades of research in the field, it is safe to say that one of the major reasons underlying the poor efficacy of anti-IGF-targeting agents is derived from an underestimation of the molecular complexity of this axis. Genetic, transcriptional, post-transcriptional and functional interactors interfere with the activity of canonical components of this axis, supporting the need for combinatorial approaches to effectively block this system. In addition, cancer cells interface with a multiplicity of factors from the extracellular compartment, which strongly affect cell destiny. In this review, we will cover novel extracellular mechanisms contributing to IGF system dysregulation and the implications of such dangerous liaisons for cancer hallmarks and responses to known and new anti-IGF drugs. A deeper understanding of both the intracellular and extracellular microenvironments might provide new impetus to better decipher the complexity of the IGF axis in cancer and provide new clues for designing novel therapeutic approaches.
Collapse
Affiliation(s)
- Caterina Mancarella
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Andrea Morrione
- Sbarro Institute for Cancer Research and Molecular Medicine and Center for Biotechnology, Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA;
| | - Katia Scotlandi
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| |
Collapse
|
18
|
Franzese O, Ancona P, Bianchi N, Aguiari G. Apoptosis, a Metabolic "Head-to-Head" between Tumor and T Cells: Implications for Immunotherapy. Cells 2024; 13:924. [PMID: 38891056 PMCID: PMC11171541 DOI: 10.3390/cells13110924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/18/2024] [Accepted: 05/22/2024] [Indexed: 06/20/2024] Open
Abstract
Induction of apoptosis represents a promising therapeutic approach to drive tumor cells to death. However, this poses challenges due to the intricate nature of cancer biology and the mechanisms employed by cancer cells to survive and escape immune surveillance. Furthermore, molecules released from apoptotic cells and phagocytes in the tumor microenvironment (TME) can facilitate cancer progression and immune evasion. Apoptosis is also a pivotal mechanism in modulating the strength and duration of anti-tumor T-cell responses. Combined strategies including molecular targeting of apoptosis, promoting immunogenic cell death, modulating immunosuppressive cells, and affecting energy pathways can potentially overcome resistance and enhance therapeutic outcomes. Thus, an effective approach for targeting apoptosis within the TME should delicately balance the selective induction of apoptosis in tumor cells, while safeguarding survival, metabolic changes, and functionality of T cells targeting crucial molecular pathways involved in T-cell apoptosis regulation. Enhancing the persistence and effectiveness of T cells may bolster a more resilient and enduring anti-tumor immune response, ultimately advancing therapeutic outcomes in cancer treatment. This review delves into the pivotal topics of this multifaceted issue and suggests drugs and druggable targets for possible combined therapies.
Collapse
Affiliation(s)
- Ornella Franzese
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy;
| | - Pietro Ancona
- Department of Translational Medicine, University of Ferrara, Via Fossato di Mortara 70, 44121 Ferrara, Italy;
| | - Nicoletta Bianchi
- Department of Translational Medicine, University of Ferrara, Via Fossato di Mortara 70, 44121 Ferrara, Italy;
| | - Gianluca Aguiari
- Department of Neuroscience and Rehabilitation, University of Ferrara, Via F. Mortara 74, 44121 Ferrara, Italy;
| |
Collapse
|
19
|
Fey RM, Nichols RA, Tran TT, Vandenbark AA, Kulkarni RP. MIF and CD74 as Emerging Biomarkers for Immune Checkpoint Blockade Therapy. Cancers (Basel) 2024; 16:1773. [PMID: 38730725 PMCID: PMC11082995 DOI: 10.3390/cancers16091773] [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] [Revised: 04/18/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024] Open
Abstract
Immune checkpoint blockade (ICB) therapy is used to treat a wide range of cancers; however, some patients are at risk of developing treatment resistance and/or immune-related adverse events (irAEs). Thus, there is a great need for the identification of reliable predictive biomarkers for response and toxicity. The cytokine MIF (macrophage migration inhibitory factor) and its cognate receptor CD74 are intimately connected with cancer progression and have previously been proposed as prognostic biomarkers for patient outcome in various cancers, including solid tumors such as malignant melanoma. Here, we assess their potential as predictive biomarkers for response to ICB therapy and irAE development. We provide a brief overview of their function and roles in the context of cancer and autoimmune disease. We also review the evidence showing that MIF and CD74 may be of use as predictive biomarkers of patient response to ICB therapy and irAE development. We also highlight that careful consideration is required when assessing the potential of serum MIF levels as a biomarker due to its reported circadian expression in human plasma. Finally, we suggest future directions for the establishment of MIF and CD74 as predictive biomarkers for ICB therapy and irAE development to guide further research in this field.
Collapse
Affiliation(s)
- Rosalyn M. Fey
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA (R.A.N.)
| | - Rebecca A. Nichols
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA (R.A.N.)
| | - Thuy T. Tran
- Yale Cancer Center, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Arthur A. Vandenbark
- Neuroimmunology Research, R&D-31, VA Portland Health Care System, Portland, OR 97239, USA
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Rajan P. Kulkarni
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA (R.A.N.)
- Cancer Early Detection Advanced Research Center (CEDAR), Portland, OR 97239, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
- Operative Care Division, U.S. Department of Veterans Affairs Portland Health Care System, Portland, OR 97239, USA
| |
Collapse
|
20
|
Uchi H. Optimal strategy in managing advanced melanoma. J Dermatol 2024; 51:324-334. [PMID: 38087810 PMCID: PMC11483965 DOI: 10.1111/1346-8138.17068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 11/01/2023] [Accepted: 11/20/2023] [Indexed: 03/05/2024]
Abstract
The advent of immune checkpoint inhibitors and combination therapy with BRAF inhibitors and MEK inhibitors has dramatically improved the prognosis of advanced melanoma. However, since acral melanoma and mucosal melanoma, which are rare in Western countries but are major subtypes of melanoma in East Asia, including Japan, have a low frequency of BRAF mutations, there are currently no treatment options other than immune checkpoint inhibitors in most such cases. Furthermore, owing to a lower tumor mutation burden, immune checkpoint inhibitors are less effective in acral and mucosal melanoma than in cutaneous melanoma. The aim of this review was to summarize the current status and future prospects for the treatment of advanced melanoma, comparing cutaneous melanoma, acral melanoma, and mucosal melanoma.
Collapse
Affiliation(s)
- Hiroshi Uchi
- Department of Dermato‐OncologyNational Hospital Organization Kyushu Cancer CenterFukuokaJapan
| |
Collapse
|
21
|
Khoo A, Boyer M, Jafri Z, Makeham T, Pham T, Khachigian LM, Floros P, Dowling E, Fedder K, Shonka D, Garneau J, O'Meara CH. Human Papilloma Virus Positive Oropharyngeal Squamous Cell Carcinoma and the Immune System: Pathogenesis, Immunotherapy and Future Perspectives. Int J Mol Sci 2024; 25:2798. [PMID: 38474047 DOI: 10.3390/ijms25052798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 02/14/2024] [Accepted: 02/16/2024] [Indexed: 03/14/2024] Open
Abstract
Oropharyngeal squamous cell carcinoma (OPSCC), a subset of head and neck squamous cell carcinoma (HNSCC), involves the palatine tonsils, soft palate, base of tongue, and uvula, with the ability to spread to adjacent subsites. Personalized treatment strategies for Human Papillomavirus-associated squamous cell carcinoma of the oropharynx (HPV+OPSCC) are yet to be established. In this article, we summarise our current understanding of the pathogenesis of HPV+OPSCC, the intrinsic role of the immune system, current ICI clinical trials, and the potential role of small molecule immunotherapy in HPV+OPSCC.
Collapse
Affiliation(s)
- A Khoo
- Department of Otolaryngology, Head & Neck Surgery, Canberra Health Services, Canberra, ACT 2601, Australia
| | - M Boyer
- Chris O'Brien Lifehouse, Camperdown, NSW 2050, Australia
| | - Z 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
| | - T Makeham
- Department of Otolaryngology, Head & Neck Surgery, Canberra Health Services, Canberra, ACT 2601, Australia
- ANU School of Medicine & Psychology, Australian National University, Canberra, ACT 0200, Australia
| | - T Pham
- Department of Otolaryngology, Head & Neck Surgery, Canberra Health Services, Canberra, ACT 2601, Australia
- ANU School of Medicine & Psychology, Australian National University, Canberra, ACT 0200, Australia
| | - L 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
| | - P Floros
- St Vincent's Hospital, 390 Victoria Street, Sydney, NSW 2010, Australia
| | - E Dowling
- Department of Otolaryngology, Head & Neck Surgery, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - K Fedder
- Department of Otolaryngology, Head & Neck Surgery, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - D Shonka
- Department of Otolaryngology, Head & Neck Surgery, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - J Garneau
- Department of Otolaryngology, Head & Neck Surgery, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - C H O'Meara
- Department of Otolaryngology, Head & Neck Surgery, Canberra Health Services, Canberra, ACT 2601, Australia
- ANU School of Medicine & Psychology, Australian National University, Canberra, ACT 0200, Australia
- Department of Otolaryngology, Head & Neck Surgery, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| |
Collapse
|
22
|
Pessino G, Scotti C, Maggi M, Immuno-Hub Consortium. Hepatocellular Carcinoma: Old and Emerging Therapeutic Targets. Cancers (Basel) 2024; 16:901. [PMID: 38473265 DOI: 10.3390/cancers16050901] [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: 01/31/2024] [Revised: 02/16/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
Liver cancer, predominantly hepatocellular carcinoma (HCC), globally ranks sixth in incidence and third in cancer-related deaths. HCC risk factors include non-viral hepatitis, alcohol abuse, environmental exposures, and genetic factors. No specific genetic alterations are unequivocally linked to HCC tumorigenesis. Current standard therapies include surgical options, systemic chemotherapy, and kinase inhibitors, like sorafenib and regorafenib. Immunotherapy, targeting immune checkpoints, represents a promising avenue. FDA-approved checkpoint inhibitors, such as atezolizumab and pembrolizumab, show efficacy, and combination therapies enhance clinical responses. Despite this, the treatment of hepatocellular carcinoma (HCC) remains a challenge, as the complex tumor ecosystem and the immunosuppressive microenvironment associated with it hamper the efficacy of the available therapeutic approaches. This review explores current and advanced approaches to treat HCC, considering both known and new potential targets, especially derived from proteomic analysis, which is today considered as the most promising approach. Exploring novel strategies, this review discusses antibody drug conjugates (ADCs), chimeric antigen receptor T-cell therapy (CAR-T), and engineered antibodies. It then reports a systematic analysis of the main ligand/receptor pairs and molecular pathways reported to be overexpressed in tumor cells, highlighting their potential and limitations. Finally, it discusses TGFβ, one of the most promising targets of the HCC microenvironment.
Collapse
Affiliation(s)
- Greta Pessino
- Unit of Immunology and General Pathology, Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Claudia Scotti
- Unit of Immunology and General Pathology, Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Maristella Maggi
- Unit of Immunology and General Pathology, Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Immuno-Hub Consortium
- Unit of Immunology and General Pathology, Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
23
|
Ouchida T, Suzuki H, Tanaka T, Kaneko MK, Kato Y. Establishment of Anti-Dog Programmed Cell Death Ligand 1 Monoclonal Antibodies for Immunohistochemistry. Monoclon Antib Immunodiagn Immunother 2024; 43:17-23. [PMID: 38237003 DOI: 10.1089/mab.2023.0014] [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: 02/28/2024] Open
Abstract
Immune checkpoint blockade therapy has shown successful clinical outcomes in multiple human cancers. In dogs, several types of tumors resemble human tumors in many respects. Therefore, several groups have developed the anti-dog programmed cell death ligand 1 (dPD-L1) monoclonal antibodies (mAbs) and showed efficacy in several canine tumors. To examine the abundance of dPD-L1 in canine tumors, anti-dPD-L1 diagnostic mAbs for immunohistochemistry are required. In this study, we immunized the peptide in the dPD-L1 intracellular domain, and established anti-dPD-L1 mAbs, L1Mab-352 (mouse IgG1, kappa), and L1Mab-354 (mouse IgG1, kappa). In enzyme-linked immunosorbent assay, L1Mab-352 and L1Mab-354 showed high-binding affinity to the dPD-L1 peptide, and the dissociation constants (KD) were determined as 6.9 × 10-10 M and 7.2 × 10-10 M, respectively. Furthermore, L1Mab-352 and L1Mab-354 were applicable for the detection of dPD-L1 in immunohistochemical analysis in paraffin-embedded dPD-L1-overexpressed cells. These results indicated that L1Mab-352 and L1Mab-354 are useful for detecting dPD-L1 in immunohistochemical analysis.
Collapse
Affiliation(s)
- Tsunenori Ouchida
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroyuki Suzuki
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tomohiro Tanaka
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Mika K Kaneko
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yukinari Kato
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
24
|
Komatsuda H, Kono M, Wakisaka R, Sato R, Inoue T, Kumai T, Takahara M. Harnessing Immunity to Treat Advanced Thyroid Cancer. Vaccines (Basel) 2023; 12:45. [PMID: 38250858 PMCID: PMC10820966 DOI: 10.3390/vaccines12010045] [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: 11/20/2023] [Revised: 12/26/2023] [Accepted: 12/29/2023] [Indexed: 01/23/2024] Open
Abstract
The incidence of thyroid cancer (TC) has increased over the past 30 years. Although differentiated thyroid cancer (DTC) has a good prognosis in most patients undergoing total thyroidectomy followed by radioiodine therapy (RAI), 5-10% of patients develop metastasis. Anaplastic thyroid cancer (ATC) has a low survival rate and few effective treatments have been available to date. Recently, tyrosine kinase inhibitors (TKIs) have been successfully applied to RAI-resistant or non-responsive TC to suppress the disease. However, TC eventually develops resistance to TKIs. Immunotherapy is a promising treatment for TC, the majority of which is considered an immune-hot malignancy. Immune suppression by TC cells and immune-suppressing cells, including tumor-associated macrophages, myeloid-derived suppressor cells, and regulatory T cells, is complex and dynamic. Negative immune checkpoints, cytokines, vascular endothelial growth factors (VEGF), and indoleamine 2,3-dioxygenase 1 (IDO1) suppress antitumor T cells. Basic and translational advances in immune checkpoint inhibitors (ICIs), molecule-targeted therapy, tumor-specific immunotherapy, and their combinations have enabled us to overcome immune suppression and activate antitumor immune cells. This review summarizes current findings regarding the immune microenvironment, immunosuppression, immunological targets, and immunotherapy for TC and highlights the potential efficacy of immunotherapy.
Collapse
Affiliation(s)
- Hiroki Komatsuda
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Asahikawa 078-8510, Japan; (H.K.); (M.K.); (R.W.); (R.S.); (T.I.); (M.T.)
| | - Michihisa Kono
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Asahikawa 078-8510, Japan; (H.K.); (M.K.); (R.W.); (R.S.); (T.I.); (M.T.)
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Risa Wakisaka
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Asahikawa 078-8510, Japan; (H.K.); (M.K.); (R.W.); (R.S.); (T.I.); (M.T.)
| | - Ryosuke Sato
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Asahikawa 078-8510, Japan; (H.K.); (M.K.); (R.W.); (R.S.); (T.I.); (M.T.)
| | - Takahiro Inoue
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Asahikawa 078-8510, Japan; (H.K.); (M.K.); (R.W.); (R.S.); (T.I.); (M.T.)
| | - Takumi Kumai
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Asahikawa 078-8510, Japan; (H.K.); (M.K.); (R.W.); (R.S.); (T.I.); (M.T.)
- Department of Innovative Head & Neck Cancer Research and Treatment, Asahikawa Medical University, Asahikawa 078-8510, Japan
| | - Miki Takahara
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Asahikawa 078-8510, Japan; (H.K.); (M.K.); (R.W.); (R.S.); (T.I.); (M.T.)
- Department of Innovative Head & Neck Cancer Research and Treatment, Asahikawa Medical University, Asahikawa 078-8510, Japan
| |
Collapse
|
25
|
Jia H, Yang H, Xiong H, Luo KQ. NK cell exhaustion in the tumor microenvironment. Front Immunol 2023; 14:1303605. [PMID: 38022646 PMCID: PMC10653587 DOI: 10.3389/fimmu.2023.1303605] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Natural killer (NK) cells kill mutant cells through death receptors and cytotoxic granules, playing an essential role in controlling cancer progression. However, in the tumor microenvironment (TME), NK cells frequently exhibit an exhausted status, which impairs their immunosurveillance function and contributes to tumor immune evasion. Emerging studies are ongoing to reveal the properties and mechanisms of NK cell exhaustion in the TME. In this review, we will briefly introduce the maturation, localization, homeostasis, and cytotoxicity of NK cells. We will then summarize the current understanding of the main mechanisms underlying NK cell exhaustion in the TME in four aspects: dysregulation of inhibitory and activating signaling, tumor cell-derived factors, immunosuppressive cells, and metabolism and exhaustion. We will also discuss the therapeutic approaches currently being developed to reverse NK cell exhaustion and enhance NK cell cytotoxicity in the TME.
Collapse
Affiliation(s)
- Hao Jia
- Faculty of Health Sciences, University of Macau, Taipa, Macao SAR, China
| | - Hongmei Yang
- Faculty of Health Sciences, University of Macau, Taipa, Macao SAR, China
| | - Huaxing Xiong
- Faculty of Health Sciences, University of Macau, Taipa, Macao SAR, China
| | - Kathy Qian Luo
- Faculty of Health Sciences, University of Macau, Taipa, Macao SAR, China
- Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macao SAR, China
| |
Collapse
|
26
|
Boutros M, Attieh F, Chartouni A, Jalbout J, Kourie HR. Beyond the Horizon: A Cutting-Edge Review of the Latest Checkpoint Inhibitors in Cancer Treatment. Cancer Invest 2023; 41:757-773. [PMID: 37795860 DOI: 10.1080/07357907.2023.2267675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 10/03/2023] [Indexed: 10/06/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have emerged as a revolutionary paradigm in oncology, offering a potent arsenal against various malignancies by harnessing the body's own immunological prowess. In a whirlwind of advancement, an abundance of new ICIs have come to light, rendering it a Herculean task for physicians to remain au courant with the rapidly evolving landscape. This comprehensive review meticulously explores the crescendo of clinical investigations and FDA approvals that have come to light during 2022 and 2023, showcasing the metamorphic impact of ICIs in cancer therapeutics. Delving into the pith of pivotal Phase 3 trials across diverse cancer types - including lung, renal, melanoma, and more - the review illuminates the significant strides made in enhancing patient outcomes, alongside the unveiling of novel ICIs that have garnered attention in the oncological community. The analysis extends to the notable presentations at the esteemed ESMO and ASCO conventions, providing a panoramic view of the contemporary advancements in ICI technology. Furthermore, the review underscores the imperative of continuous exploration in overcoming the extant challenges, such as the quest for reliable predictive biomarkers and the optimization of combinatorial strategies to surmount resistance and augment therapeutic efficacy. Through a holistic lens, this article elucidates the monumental impact of ICIs, marking a significant epoch in the odyssey towards rendering cancer a conquerable adversary.
Collapse
Affiliation(s)
- Marc Boutros
- Faculty of Medicine, Saint Joseph University of Beirut, Beirut, Lebanon
| | - Fouad Attieh
- Faculty of Medicine, Saint Joseph University of Beirut, Beirut, Lebanon
| | - Antoine Chartouni
- Faculty of Medicine, Saint Joseph University of Beirut, Beirut, Lebanon
| | - Johnny Jalbout
- Faculty of Medicine, Saint Joseph University of Beirut, Beirut, Lebanon
| | - Hampig Raphaël Kourie
- Department of Hematology-Oncology, Faculty of Medicine, Saint-Joseph University of Beirut, Beirut, Lebanon
| |
Collapse
|
27
|
Wang Y, Lu L, Ling C, Zhang P, Han R. Potential of Dietary HDAC2i in Breast Cancer Patients Receiving PD-1/PD-L1 Inhibitors. Nutrients 2023; 15:3984. [PMID: 37764768 PMCID: PMC10537481 DOI: 10.3390/nu15183984] [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: 08/02/2023] [Revised: 09/09/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Breast cancer (BC) is a lethal malignancy with high morbidity and mortality but lacks effective treatments thus far. Despite the introduction of immune checkpoint inhibitors (ICIs) (including PD-1/PD-L1 inhibitors), durable and optimal clinical benefits still remain elusive for a considerable number of BC patients. To break through such a dilemma, novel ICI-based combination therapy has been explored for enhancing the therapeutic effect. Recent evidence has just pointed out that the HDAC2 inhibitor (HDAC2i), which has been proven to exhibit an anti-cancer effect, can act as a sensitizer for ICIs therapy. Simultaneously, dietary intervention, as a crucial supportive therapy, has been reported to provide ingredients containing HDAC2 inhibitory activity. Thus, the novel integration of dietary intervention with ICIs therapy may offer promising possibilities for improving treatment outcomes. In this study, we first conducted the differential expression and prognostic analyses of HDAC2 and BC patients using the GENT2 and Kaplan-Meier plotter platform. Then, we summarized the potential diet candidates for such an integrated therapeutic strategy. This article not only provides a whole new therapeutic strategy for an HDAC2i-containing diet combined with PD-1/PD-L1 inhibitors for BC treatment, but also aims to ignite enthusiasm for exploring this field.
Collapse
Affiliation(s)
- Yuqian Wang
- Department of Chinese Medicine Oncology, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
- Department of Chinese Medicine, Naval Medical University, Shanghai 200433, China
| | - Lingeng Lu
- Department of Chronic Disease Epidemiology, Yale School of Public Health, Yale University, 60 College Street, New Haven, CT 06520, USA
- School of Medicine, Center for Biomedical Data Science, Yale University, 60 College Street, New Haven, CT 06520, USA
- Yale Cancer Center, Yale University, 60 College Street, New Haven, CT 06520, USA
| | - Changquan Ling
- Department of Chinese Medicine Oncology, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
- Department of Chinese Medicine, Naval Medical University, Shanghai 200433, China
| | - Ping Zhang
- Center for Integrative Conservation, Yunnan Key Laboratory for the Conservation of Tropical Rainforests and Asian Elephants, Xishuangbanna Tropical Botanical Garden, Xishuangbanna 666303, China
| | - Rui Han
- Department of Chinese Medicine Oncology, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
- Department of Chinese Medicine, Naval Medical University, Shanghai 200433, China
- Department of Chronic Disease Epidemiology, Yale School of Public Health, Yale University, 60 College Street, New Haven, CT 06520, USA
- School of Medicine, Center for Biomedical Data Science, Yale University, 60 College Street, New Haven, CT 06520, USA
- Yale Cancer Center, Yale University, 60 College Street, New Haven, CT 06520, USA
- Department of Oncology, The First Hospital Affiliated to Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| |
Collapse
|