201
|
Zhou X, Peng M, He Y, Peng J, Zhang X, Wang C, Xia X, Song W. CXC Chemokines as Therapeutic Targets and Prognostic Biomarkers in Skin Cutaneous Melanoma Microenvironment. Front Oncol 2021; 11:619003. [PMID: 33767987 PMCID: PMC7985846 DOI: 10.3389/fonc.2021.619003] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 01/22/2021] [Indexed: 02/06/2023] Open
Abstract
Background Skin Cutaneous Melanoma (SKCM) is a tumor of the epidermal melanocytes induced by gene activation or mutation. It is the result of the interaction between genetic, constitutional, and environmental factors. SKCM is highly aggressive and is the most threatening skin tumor. The incidence of the disease is increasing year by year, and it is the main cause of death in skin tumors around the world. CXC chemokines in the tumor microenvironment can regulate the transport of immune cells and the activity of tumor cells, thus playing an anti-tumor immunological role and affecting the prognosis of patients. However, the expression level of CXC chemokine in SKCM and its effect on prognosis are still unclear. Method Oncomine, UALCAN, GEPIA, STRING, GeneMANIA, cBioPortal, TIMER, TRRUST, DAVID 6.8, and Metascape were applied in our research. Result The transcription of CXCL1, CXCL5, CXCL8, CXCL9, CXCL10, and CXCL13 in SKCM tissues were significantly higher than those in normal tissues. The pathological stage of SKCM patients is closely related to the expression of CXCL4, CXCL9, CXCL10, CXCL11, CXCL12, and CXCL13. The prognosis of SKCM patients with low transcription levels of CXCL4, CXCL9, CXCL10, CXCL11, and CXCL13 is better. The differential expression of CXC chemokines is mainly associated with inflammatory response, immune response, and cytokine mediated signaling pathways. Our data indicate that the key transcription factors of CXC chemokines are RELA, NF-κB1 and SP1. The targets of CXC chemokines are mainly LCK, LYN, SYK, MAPK2, MAPK12, and ART. The relationship between CXC chemokine expression and immune cell infiltration in SKCM was closed. Conclusions Our research provides a basis for screening SKCM biomarkers, predicting prognosis, and choosing immunotherapy.
Collapse
Affiliation(s)
- Xuezhi Zhou
- Hunan Key Laboratory of Ophthalmology, Eye Center of Xiangya Hospital, Central South University, Changsha, China
| | - Manjuan Peng
- Hunan Key Laboratory of Ophthalmology, Eye Center of Xiangya Hospital, Central South University, Changsha, China
| | - Ye He
- Hunan Key Laboratory of Ophthalmology, Eye Center of Xiangya Hospital, Central South University, Changsha, China
| | - Jingjie Peng
- Hunan Key Laboratory of Ophthalmology, Eye Center of Xiangya Hospital, Central South University, Changsha, China
| | - Xuan Zhang
- Hunan Key Laboratory of Ophthalmology, Eye Center of Xiangya Hospital, Central South University, Changsha, China
| | - Chao Wang
- Hunan Key Laboratory of Ophthalmology, Eye Center of Xiangya Hospital, Central South University, Changsha, China
| | - Xiaobo Xia
- Hunan Key Laboratory of Ophthalmology, Eye Center of Xiangya Hospital, Central South University, Changsha, China
| | - Weitao Song
- Hunan Key Laboratory of Ophthalmology, Eye Center of Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
202
|
Huang X, Tang T, Zhang G, Liang T. Identification of tumor antigens and immune subtypes of cholangiocarcinoma for mRNA vaccine development. Mol Cancer 2021; 20:50. [PMID: 33685460 PMCID: PMC7938044 DOI: 10.1186/s12943-021-01342-6] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 02/23/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The mRNA-based cancer vaccine has been considered a promising strategy and the next hotspot in cancer immunotherapy. However, its application on cholangiocarcinoma remains largely uncharacterized. This study aimed to identify potential antigens of cholangiocarcinoma for development of anti-cholangiocarcinoma mRNA vaccine, and determine immune subtypes of cholangiocarcinoma for selection of suitable patients from an extremely heterogeneous population. METHODS Gene expression profiles and corresponding clinical information were collected from GEO and TCGA, respectively. cBioPortal was used to visualize and compare genetic alterations. GEPIA2 was used to calculate the prognostic index of the selected antigens. TIMER was used to visualize the correlation between the infiltration of antigen-presenting cells and the expression of the identified antigens. Consensus clustering analysis was performed to identify the immune subtypes. Graph learning-based dimensionality reduction analysis was conducted to visualize the immune landscape of cholangiocarcinoma. RESULTS Three tumor antigens, such as CD247, FCGR1A, and TRRAP, correlated with superior prognoses and infiltration of antigen-presenting cells were identified in cholangiocarcinoma. Cholangiocarcinoma patients were stratified into two immune subtypes characterized by differential molecular, cellular and clinical features. Patients with the IS1 tumor had immune "hot" and immunosuppressive phenotype, whereas those with the IS2 tumor had immune "cold" phenotype. Interestingly, patients with the IS2 tumor had a superior survival than those with the IS1 tumor. Furthermore, distinct expression of immune checkpoints and immunogenic cell death modulators was observed between different immune subtype tumors. Finally, the immune landscape of cholangiocarcinoma revealed immune cell components in individual patient. CONCLUSIONS CD247, FCGR1A, and TRRAP are potential antigens for mRNA vaccine development against cholangiocarcinoma, specifically for patients with IS2 tumors. Therefore, this study provides a theoretical basis for the anti-cholangiocarcinoma mRNA vaccine and defines suitable patients for vaccination.
Collapse
Affiliation(s)
- Xing Huang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China.,Innovation Center for the Study of Pancreatic Diseases, Hangzhou, 310003, Zhejiang Province, China.,Cancer Center, Zhejiang University, Hangzhou, 310058, Zhejiang, China.,Research Center for Healthcare Data Science, Zhejiang Lab, Hangzhou, 310003, Zhejiang, China
| | - Tianyu Tang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China.,Innovation Center for the Study of Pancreatic Diseases, Hangzhou, 310003, Zhejiang Province, China.,Cancer Center, Zhejiang University, Hangzhou, 310058, Zhejiang, China.,Research Center for Healthcare Data Science, Zhejiang Lab, Hangzhou, 310003, Zhejiang, China
| | - Gang Zhang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China.,Innovation Center for the Study of Pancreatic Diseases, Hangzhou, 310003, Zhejiang Province, China.,Cancer Center, Zhejiang University, Hangzhou, 310058, Zhejiang, China.,Research Center for Healthcare Data Science, Zhejiang Lab, Hangzhou, 310003, Zhejiang, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China. .,Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China. .,Innovation Center for the Study of Pancreatic Diseases, Hangzhou, 310003, Zhejiang Province, China. .,Cancer Center, Zhejiang University, Hangzhou, 310058, Zhejiang, China. .,Research Center for Healthcare Data Science, Zhejiang Lab, Hangzhou, 310003, Zhejiang, China.
| |
Collapse
|
203
|
The Current Landscape of Immune Checkpoint Blockade in Metastatic Lung Squamous Cell Carcinoma. Molecules 2021; 26:molecules26051392. [PMID: 33807509 PMCID: PMC7961875 DOI: 10.3390/molecules26051392] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 02/21/2021] [Accepted: 03/02/2021] [Indexed: 02/06/2023] Open
Abstract
In addition to surgery, chemotherapy, radiotherapy, and targeted therapy, immunotherapy has emerged as a standard pillar of cancer treatment. Immune checkpoint inhibitors (ICIs) such as targeting programmed death-1/programmed death ligand 1 (PD-1/PD-L1) and cytotoxic T lymphocyte antigen 4 (CTLA-4) have been integrated into standard-of-care regimens for patients with advanced lung squamous cell carcinoma (LUSC), who were previously limited by the lack of treatment options. Atezolizumab, durvalumab, nivolumab, and pembrolizumab are all currently used as part of standard-of-care treatment for different stages of lung cancer. Recent successes and failures of immune checkpoint blockade-based combination therapies have provided significant insights into implementing combination strategies in LUSC. Therefore, there is an urgent need to correctly select patients who are more likely to respond to immunotherapy and understand the mechanisms of primary or acquired resistance. In this review, we aim at summarizing the emerging clinical data on the promise and challenge of ICIs, discussing the unmet need of potential biomarkers for predicting response or resistance to immunotherapy, and providing an overview of the current immune landscape and future directions in advanced LUSC.
Collapse
|
204
|
Glorieux C, Cui L, Zeng P, Xia X, Huang P. Diverse effects of chemotherapeutic agents on immune cell function and implications in immunochemotherapy. Cancer Commun (Lond) 2021; 41:432-435. [PMID: 33650790 PMCID: PMC8118588 DOI: 10.1002/cac2.12139] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 12/05/2020] [Accepted: 01/19/2021] [Indexed: 12/16/2022] Open
Affiliation(s)
- Christophe Glorieux
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, P. R. China
| | - Lei Cui
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, P. R. China
| | - Peiting Zeng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, P. R. China
| | - Xiaojun Xia
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, P. R. China
| | - Peng Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, P. R. China.,Metabolic Innovation Center, Sun Yat-sen University, Guangzhou, Guangdong, 510080, P. R. China
| |
Collapse
|
205
|
Zhang Y, He L, Sadagopan A, Ma T, Dotti G, Wang Y, Zheng H, Gao X, Wang D, DeLeo AB, Fan S, Sun R, Yu L, Zhang L, Wang G, Ferrone S, Wang X. Targeting Radiation-Resistant Prostate Cancer Stem Cells by B7-H3 CAR T Cells. Mol Cancer Ther 2021; 20:577-588. [PMID: 33653946 PMCID: PMC7952034 DOI: 10.1158/1535-7163.mct-20-0446] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 11/13/2020] [Accepted: 01/04/2021] [Indexed: 01/16/2023]
Abstract
Radiotherapy (RT) is a key treatment for prostate cancer. However, RT resistance can contribute to treatment failure. Prostate cancer stem cells (PCSCs) are radioresistant. We recently found that fractionated irradiation (FIR) upregulates expression of the immune checkpoint B7-H3 (CD276) on PCSCs and bulk cells in each prostate cancer cell line tested. These findings prompted us to investigate whether B7-H3 targeting chimeric antigen receptor (CAR) T cells, which may abrogate function of an immune checkpoint and mediate lysis of targeted cells, can target RT-resistant PCSCs in vitro and in vivo. B7-H3 expression is naturally higher on PCSCs than bulk prostate cancer cells and cytotoxicity of B7-H3 CAR T cells to PCSCs is more potent than to bulk prostate cancer cells. Furthermore, FIR significantly upregulates B7-H3 expression on PCSCs and bulk prostate cancer cells. The duration of FIR or single-dose irradiation-induced further upregulation of B7-H3 on bulk prostate cancer cells and PCSCs lasts for up to 3 days. B7-H3 CAR T-cell cytotoxicity against FIR-resistant PCSCs at a low effector to target ratio of 1:1 was assessed by flow cytometry and sphere formation assays. Further upregulation of B7-H3 expression by FIR made PCSCs even more sensitive to B7-H3 CAR T-cell-mediated killing. Consequently, the FIR and B7-H3 CAR T-cell therapy combination is much more effective than FIR or CAR T cells alone in growth inhibition of hormone-insensitive prostate cancer xenografts in immunodeficient mice. Our work provides a sound basis for further development of this unique combinatorial model of RT and B7-H3 CAR T-cell therapy for prostate cancer. SIGNIFICANCE: We demonstrate that FIR significantly upregulates B7-H3 expression by RT-resistant PCSCs and bulk cells; cytotoxicity of B7-H3 CAR T cells to FIR-treated PCSCs is potent and results in significantly improved antitumor efficacy in mice.
Collapse
Affiliation(s)
- Yida Zhang
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Lile He
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ananthan Sadagopan
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Tao Ma
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Gianpietro Dotti
- Lineberger Comprehensive Cancer Center and Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina
| | - Yufeng Wang
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Hui Zheng
- Biostatistics Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Xin Gao
- Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Dian Wang
- Department of Radiation Oncology, Rush University Medical Center, Chicago, Illinois
| | - Albert B DeLeo
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Song Fan
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ruochuan Sun
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ling Yu
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Liyuan Zhang
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Gongxian Wang
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Soldano Ferrone
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Xinhui Wang
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
206
|
Raza F, Zafar H, Zhang S, Kamal Z, Su J, Yuan W, Mingfeng Q. Recent Advances in Cell Membrane-Derived Biomimetic Nanotechnology for Cancer Immunotherapy. Adv Healthc Mater 2021; 10:e2002081. [PMID: 33586322 DOI: 10.1002/adhm.202002081] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/13/2021] [Indexed: 12/17/2022]
Abstract
Immunotherapy will significantly impact the standard of care in cancer treatment. Recent advances in nanotechnology can improve the efficacy of cancer immunotherapy. However, concerns regarding efficiency of cancer nanomedicine, complex tumor microenvironment, patient heterogeneity, and systemic immunotoxicity drive interest in more novel approaches to be developed. For this purpose, biomimetic nanoparticles are developed to make innovative changes in the delivery and biodistribution of immunotherapeutics. Biomimetic nanoparticles have several advantages that can advance the clinical efficacy of cancer immunotherapy. Thus there is a greater push toward the utilization of biomimetic nanotechnology for developing effective cancer immunotherapeutics that demonstrate increased specificity and potency. The recent works and state-of-the-art strategies for anti-tumor immunotherapeutics are highlighted here, and particular emphasis has been given to the applications of cell-derived biomimetic nanotechnology for cancer immunotherapy.
Collapse
Affiliation(s)
- Faisal Raza
- School of Pharmacy Shanghai Jiao Tong University Shanghai 200240 P. R. China
| | - Hajra Zafar
- School of Pharmacy Shanghai Jiao Tong University Shanghai 200240 P. R. China
| | - Shulei Zhang
- School of Pharmacy Shanghai Jiao Tong University Shanghai 200240 P. R. China
| | - Zul Kamal
- School of Pharmacy Shanghai Jiao Tong University Shanghai 200240 P. R. China
- Department of Pharmacy Shaheed Benazir Bhutto University Sheringal Dir (Upper) Khyber Pakhtunkhwa 18000 Pakistan
| | - Jing Su
- School of Pharmacy Shanghai Jiao Tong University Shanghai 200240 P. R. China
| | - Wei‐En Yuan
- School of Pharmacy Shanghai Jiao Tong University Shanghai 200240 P. R. China
| | - Qiu Mingfeng
- School of Pharmacy Shanghai Jiao Tong University Shanghai 200240 P. R. China
| |
Collapse
|
207
|
Yuan J, Ye D, Chen S, Chen H. Therapeutic ultrasound-enhanced immune checkpoint inhibitor therapy. FRONTIERS IN PHYSICS 2021; 9:636985. [PMID: 37994329 PMCID: PMC10664841 DOI: 10.3389/fphy.2021.636985] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2023]
Abstract
Immune checkpoint inhibitors (ICIs) are designed to reinvigorate antitumor immune responses by interrupting inhibitory signaling pathways and promoting the immune-mediated elimination of malignant cells. Although ICI therapy has transformed the landscape of cancer treatment, only a subset of patients achieve a complete response. Focused ultrasound (FUS) is a noninvasive, nonionizing, deep penetrating focal therapy that has great potential to improve the efficacy of ICIs in solid tumors. Five FUS modalities have been incorporated with ICIs to explore their antitumor effects in preclinical studies, namely, high-intensity focused ultrasound (HIFU) thermal ablation, HIFU hyperthermia, HIFU mechanical ablation, ultrasound-targeted microbubble destruction (UTMD), and sonodynamic therapy (SDT). The enhancement of the antitumor immune responses by these FUS modalities demonstrates the great promise of FUS as a transformative cancer treatment modality to improve ICI therapy. Here, this review summarizes these emerging applications of FUS modalities in combination with ICIs. It discusses each FUS modality, the experimental protocol for each combination strategy, the induced immune effects, and therapeutic outcomes.
Collapse
Affiliation(s)
- Jinyun Yuan
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO 63130, USA
| | - Dezhuang Ye
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO 63130, USA
| | - Si Chen
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO 63130, USA
| | - Hong Chen
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO 63130, USA
- Department of Radiation Oncology, Washington University School of Medicine, Saint Louis, MO 63108, USA
| |
Collapse
|
208
|
TIGIT/CD226 Axis Regulates Anti-Tumor Immunity. Pharmaceuticals (Basel) 2021; 14:ph14030200. [PMID: 33670993 PMCID: PMC7997242 DOI: 10.3390/ph14030200] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/23/2021] [Accepted: 02/24/2021] [Indexed: 02/07/2023] Open
Abstract
Tumors escape immune surveillance by inducing various immunosuppressive pathways, including the activation of inhibitory receptors on tumor-infiltrating T cells. While monoclonal antibodies (mAbs) blocking programmed cell death 1 (PD-1), programmed death-ligand 1 (PD-L1), and cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) have been approved for multiple cancer indications, only a subset of patients benefit from immune checkpoint blockade therapies, highlighting the need for additional approaches. Therefore, the identification of new target molecules acting in distinct or complementary pathways in monotherapy or combination therapy with PD-1/PD-L1 blockade is gaining immense interest. T cell immunoreceptor with Ig and immunoreceptor tyrosine-based inhibitory motif (ITIM) domains (TIGIT) has received considerable attention in cancer immunotherapy. Recently, anti-TIGIT mAb (tiragolumab) has demonstrated promising clinical efficacy in non-small cell lung cancer treatment when combined with an anti-PD-L1 drug (Tecentriq), leading to phase III trial initiation. TIGIT is expressed mainly on T and natural killer cells; it functions as an inhibitory checkpoint receptor, thereby limiting adaptive and innate immunity. CD226 competes for binding with the same ligands with TIGIT but delivers a positive stimulatory signal to the immune cells. This review discusses the recent discoveries regarding the roles of TIGIT and CD226 in immune cell function and their potential application in cancer immunotherapy.
Collapse
|
209
|
Tang T, Huang X, Zhang G, Hong Z, Bai X, Liang T. Advantages of targeting the tumor immune microenvironment over blocking immune checkpoint in cancer immunotherapy. Signal Transduct Target Ther 2021; 6:72. [PMID: 33608497 PMCID: PMC7896069 DOI: 10.1038/s41392-020-00449-4] [Citation(s) in RCA: 291] [Impact Index Per Article: 72.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 10/31/2020] [Accepted: 12/07/2020] [Indexed: 12/12/2022] Open
Abstract
Despite great success in cancer immunotherapy, immune checkpoint-targeting drugs are not the most popular weapon in the armory of cancer therapy. Accumulating evidence suggests that the tumor immune microenvironment plays a critical role in anti-cancer immunity, which may result in immune checkpoint blockade therapy being ineffective, in addition to other novel immunotherapies in cancer patients. In the present review, we discuss the deficiencies of current cancer immunotherapies. More importantly, we highlight the critical role of tumor immune microenvironment regulators in tumor immune surveillance, immunological evasion, and the potential for their further translation into clinical practice. Based on their general targetability in clinical therapy, we believe that tumor immune microenvironment regulators are promising cancer immunotherapeutic targets. Targeting the tumor immune microenvironment, alone or in combination with immune checkpoint-targeting drugs, might benefit cancer patients in the future.
Collapse
Affiliation(s)
- Tianyu Tang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, 310003, Hangzhou, Zhejiang, China
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 310003, Hangzhou, Zhejiang, China
- Innovation Center for the Study of Pancreatic Diseases, Zhejiang Province, 310003, Hangzhou, Zhejiang, China
- Zhejiang University Cancer Center, 310003, Hangzhou, Zhejiang, China
| | - Xing Huang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, 310003, Hangzhou, Zhejiang, China.
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 310003, Hangzhou, Zhejiang, China.
- Innovation Center for the Study of Pancreatic Diseases, Zhejiang Province, 310003, Hangzhou, Zhejiang, China.
- Zhejiang University Cancer Center, 310003, Hangzhou, Zhejiang, China.
| | - Gang Zhang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, 310003, Hangzhou, Zhejiang, China
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 310003, Hangzhou, Zhejiang, China
- Innovation Center for the Study of Pancreatic Diseases, Zhejiang Province, 310003, Hangzhou, Zhejiang, China
- Zhejiang University Cancer Center, 310003, Hangzhou, Zhejiang, China
| | - Zhengtao Hong
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, 310003, Hangzhou, Zhejiang, China
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 310003, Hangzhou, Zhejiang, China
- Innovation Center for the Study of Pancreatic Diseases, Zhejiang Province, 310003, Hangzhou, Zhejiang, China
- Zhejiang University Cancer Center, 310003, Hangzhou, Zhejiang, China
| | - Xueli Bai
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, 310003, Hangzhou, Zhejiang, China
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 310003, Hangzhou, Zhejiang, China
- Innovation Center for the Study of Pancreatic Diseases, Zhejiang Province, 310003, Hangzhou, Zhejiang, China
- Zhejiang University Cancer Center, 310003, Hangzhou, Zhejiang, China
| | - Tingbo Liang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, 310003, Hangzhou, Zhejiang, China.
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 310003, Hangzhou, Zhejiang, China.
- Innovation Center for the Study of Pancreatic Diseases, Zhejiang Province, 310003, Hangzhou, Zhejiang, China.
- Zhejiang University Cancer Center, 310003, Hangzhou, Zhejiang, China.
| |
Collapse
|
210
|
Zhou Z, Xie X, Wang X, Zhang X, Li W, Sun T, Cai Y, Wu J, Dang C, Zhang H. Correlations Between Tumor Mutation Burden and Immunocyte Infiltration and Their Prognostic Value in Colon Cancer. Front Genet 2021; 12:623424. [PMID: 33664769 PMCID: PMC7921807 DOI: 10.3389/fgene.2021.623424] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/21/2021] [Indexed: 12/12/2022] Open
Abstract
Background Colon cancer has a huge incidence and mortality worldwide every year. Immunotherapy could be a new therapeutic option for patients with advanced colon cancer. Tumor mutation burden (TMB) and immune infiltration are considered critical in immunotherapy but their characteristics in colon cancer are still controversial. Methods The somatic mutation, transcriptome, and clinical data of patients with colon cancer were obtained from the TCGA database. Patients were divided into low or high TMB groups using the median TMB value. Somatic mutation landscape, differentially expressed genes, and immune-related hub genes, Gene Ontology and KEGG, gene set enrichment, and immune infiltration analyses were investigated between the two TMB groups. Univariate and multivariate Cox analyses were utilized to construct a prognostic gene signature. The differences in immune infiltration, and the expression of HLA-related genes and checkpoint genes were investigated between the two immunity groups based on single sample gene set enrichment analysis. Finally, a nomogram of the prognostic prediction model integrating TMB, immune infiltration, and clinical parameters was established. Calibration plots and receiver operating characteristic curves (ROC) were drawn, and the C-index was calculated to assess the predictive ability. Results Missense mutations and single nucleotide polymorphisms were the major variant characteristics in colon cancer. The TMB level showed significant differences in N stage, M stage, pathological stage, and immune infiltration. CD8+ T cells, activated memory CD4+ T cells, activated NK cells, and M1 macrophages infiltrated more in the high-TMB group. The antigen processing and presentation signaling pathway was enriched in the high-TMB group. Two immune related genes (CHGB and SCT) were identified to be correlated with colon cancer survival (HR = 1.39, P = 0.01; HR = 1.26, P = 0.02, respectively). Notably, the expression of SCT was identified as a risk factor in the immune risk model, in which high risk patients showed poorer survival (P = 0.04). High immunity status exhibited significant correlations with immune response pathways, HLA-related genes, and immune checkpoint genes. Finally, including nine factors, our nomogram prediction model showed better calibration (C-index = 0.764) and had an AUC of 0.737. Conclusion In this study, we investigated the patterns and prognostic roles of TMB and immune infiltration in colon cancer, which provided new insights into the tumor microenvironment and immunotherapies and the development of a novel nomogram prognostic prediction model for patients with colon cancer.
Collapse
Affiliation(s)
- Zhangjian Zhou
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xin Xie
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xuan Wang
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xin Zhang
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Wenxin Li
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Tuanhe Sun
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yifan Cai
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jianhua Wu
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Chengxue Dang
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hao Zhang
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
211
|
Modulation of the Vascular-Immune Environment in Metastatic Cancer. Cancers (Basel) 2021; 13:cancers13040810. [PMID: 33671981 PMCID: PMC7919367 DOI: 10.3390/cancers13040810] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 12/12/2022] Open
Abstract
Advanced metastatic cancer is rarely curable. While immunotherapy has changed the oncological landscape profoundly, cure in metastatic disease remains the exception. Tumor blood vessels are crucial regulators of tumor perfusion, immune cell influx and metastatic dissemination. Indeed, vascular hyperpermeability is a key feature of primary tumors, the pre-metastatic niche in host tissue and overt metastases at secondary sites. Combining anti-angiogenesis and immune therapies may therefore unlock synergistic effects by inducing a stabilized vascular network permissive for effector T cell trafficking and function. However, anti-angiogenesis therapies, as currently applied, are hampered by intrinsic or adaptive resistance mechanisms at primary and distant tumor sites. In particular, heterogeneous vascular and immune environments which can arise in metastatic lesions of the same individual pose significant challenges for currently approved drugs. Thus, more consideration needs to be given to tailoring new combinations of vascular and immunotherapies, including dosage and timing regimens to specific disease microenvironments.
Collapse
|
212
|
Targeting IL-21 to tumor-reactive T cells enhances memory T cell responses and anti-PD-1 antibody therapy. Nat Commun 2021; 12:951. [PMID: 33574265 PMCID: PMC7878483 DOI: 10.1038/s41467-021-21241-0] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 01/15/2021] [Indexed: 12/26/2022] Open
Abstract
T cell rejuvenation by PD-1/PD-L1 blockade, despite emerging as a highly promising therapy for advanced cancers, is only beneficial for a minority of treated patients. There is evidence that a lack of efficient T cell activation may be responsible for the failure. Here, we demonstrate that IL-21 can be targeted to tumor-reactive T cells by fusion of IL-21 to anti-PD-1 antibody. To our surprise, the fusion protein PD-1Ab21 promotes the generation of memory stem T cells (TSCM) with enhanced cell proliferation. PD-1Ab21 treatment show potent antitumor effects in established tumor-bearing mice accompanied with an increased frequency of TSCM and robust expansion of tumor-specific CD8+ T cells with a memory phenotype, and is superior to a combination of PD-1 blockade and IL-21 infusion. Therefore, we have developed a potential strategy to improve the therapeutic effects of immune checkpoint blockade by simultaneously targeting cytokines to tumor-reactive T cells.
Collapse
|
213
|
Oncolytic vaccinia virus induces a novel phenotype of CD8 + effector T cells characterized by high ICOS expression. MOLECULAR THERAPY-ONCOLYTICS 2021; 20:422-432. [PMID: 33665362 PMCID: PMC7900640 DOI: 10.1016/j.omto.2021.01.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 01/26/2021] [Indexed: 12/21/2022]
Abstract
Characterization of the intratumoral immune status is important for developing immunotherapies and evaluating their antitumor effectiveness. CD8+ T cells are one of the most important cell types that directly and indirectly contribute to antitumor efficacy by releasing cytolytic molecules and inflammatory cytokines in the tumor microenvironment. Previously, we engineered a tumor-selective oncolytic vaccinia virus that encodes interleukin-7 (IL-7) and IL-12 and demonstrated its usefulness as an agent for in situ vaccination against tumors, with data showing that antitumor efficacy was reliant upon CD8+ T cells recruited by viral treatment. Here, we investigated the phenotypic changes in intratumoral CD8+ T cells caused by this oncolytic virus and found increased expression of inducible co-stimulator (ICOS) in PD-1-CD8+ T cells. Unlike previously reported ICOS+CD8+ T cells, a subset of ICOS+PD-1-CD8+ T cells showed effector function characterized by granzyme B expression. ICOS expression was induced by the backbone virus, which did not encode any immune transgenes and was independent of upregulation of the type I interferon pathway. Not only did we identify a novel effector cell subset characterized by ICOS expression, but our findings also shed light on a potential unknown aspect of the mechanism of oncolytic vaccinia virotherapy.
Collapse
|
214
|
Richardson JR, Schöllhorn A, Gouttefangeas C, Schuhmacher J. CD4+ T Cells: Multitasking Cells in the Duty of Cancer Immunotherapy. Cancers (Basel) 2021; 13:596. [PMID: 33546283 PMCID: PMC7913359 DOI: 10.3390/cancers13040596] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/24/2021] [Accepted: 01/29/2021] [Indexed: 12/22/2022] Open
Abstract
Cancer immunotherapy activates the immune system to specifically target malignant cells. Research has often focused on CD8+ cytotoxic T cells, as those have the capacity to eliminate tumor cells after specific recognition upon TCR-MHC class I interaction. However, CD4+ T cells have gained attention in the field, as they are not only essential to promote help to CD8+ T cells, but are also able to kill tumor cells directly (via MHC-class II dependent recognition) or indirectly (e.g., via the activation of other immune cells like macrophages). Therefore, immunotherapy approaches have shifted from only stimulating CD8+ T cells to targeting and assessing both, CD4+ and CD8+ T cell subsets. Here, we discuss the various subsets of CD4+ T cells, their plasticity and functionality, their relevance in the antitumor immune response in patients affected by cancer, and their ever-growing role in therapeutic approaches for human cancer.
Collapse
Affiliation(s)
- Jennifer R. Richardson
- Department of Immunology, Institute for Cell Biology, University of Tübingen, 72076 Tübingen, Germany; (J.R.R.); (A.S.); (J.S.)
| | - Anna Schöllhorn
- Department of Immunology, Institute for Cell Biology, University of Tübingen, 72076 Tübingen, Germany; (J.R.R.); (A.S.); (J.S.)
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, 72076 Tübingen, Germany
| | - Cécile Gouttefangeas
- Department of Immunology, Institute for Cell Biology, University of Tübingen, 72076 Tübingen, Germany; (J.R.R.); (A.S.); (J.S.)
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, 72076 Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) Partner Site Tübingen, 72076 Tübingen, Germany
| | - Juliane Schuhmacher
- Department of Immunology, Institute for Cell Biology, University of Tübingen, 72076 Tübingen, Germany; (J.R.R.); (A.S.); (J.S.)
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, 72076 Tübingen, Germany
| |
Collapse
|
215
|
Anderson TS, Wooster AL, La-Beck NM, Saha D, Lowe DB. Antibody-drug conjugates: an evolving approach for melanoma treatment. Melanoma Res 2021; 31:1-17. [PMID: 33165241 DOI: 10.1097/cmr.0000000000000702] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Melanoma continues to be an aggressive and deadly form of skin cancer while therapeutic options are continuously developing in an effort to provide long-term solutions for patients. Immunotherapeutic strategies incorporating antibody-drug conjugates (ADCs) have seen varied levels of success across tumor types and represent a promising approach for melanoma. This review will explore the successes of FDA-approved ADCs to date compared to the ongoing efforts of melanoma-targeting ADCs. The challenges and opportunities for future therapeutic development are also examined to distinguish how ADCs may better impact individuals with malignancies such as melanoma.
Collapse
Affiliation(s)
| | | | - Ninh M La-Beck
- Departments of Immunotherapeutics and Biotechnology
- Pharmacy Practice, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, Texas, USA
| | | | - Devin B Lowe
- Departments of Immunotherapeutics and Biotechnology
| |
Collapse
|
216
|
Ooki A, Shinozaki E, Yamaguchi K. Immunotherapy in Colorectal Cancer: Current and Future Strategies. J Anus Rectum Colon 2021; 5:11-24. [PMID: 33537496 PMCID: PMC7843143 DOI: 10.23922/jarc.2020-064] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 09/30/2020] [Indexed: 12/24/2022] Open
Abstract
Despite the recent advances in the systemic treatment of metastatic colorectal cancer (mCRC), prognostic outcomes have remained to be poor. Thus, what is needed is an innovative treatment approach. Immune checkpoint inhibitors (ICIs) targeting programmed death-1 (PD-1) and anti-programmed cell death ligand 1 (PD-L1) have exhibited a durable response and dominated the treatment of various tumor types. However, in mCRC, the clinical benefit is limited in patients with deficient mismatch repair (dMMR)/high levels of microsatellite instability (MSI-H), comprising approximately 5% of mCRC cases, and some do not respond to ICI treatment. Thus, further research is needed to identify predictive biomarkers. The most urgent need is developing effective immunotherapy for patients with proficient mismatch repair (pMMR)/microsatellite stable (MSS) cancer, which comprises 95% of mCRC cases. Tumors with the pMMR/MSS phenotype often exhibit a lower tumor mutation burden and fewer tumor-infiltrating lymphocytes than dMMR/MSI-H, leading to immune tolerance and evasion in the tumor microenvironment. Therefore, a number of investigative studies aimed at overcoming tumor resistance in current immunotherapy approaches are underway. A better understanding on the complexity and diversity of the immune system's functioning within the tumor microenvironment will increase the potential for developing predictive biomarkers and novel therapeutic strategies to potentiate anti-tumor immunity in patients with mCRC. In this review, we summarize the most recent advances in immunotherapy based on the findings of pivotal clinical trials for patients with mCRC, highlighting potent therapeutic approaches and predictive biomarkers.
Collapse
Affiliation(s)
- Akira Ooki
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Eiji Shinozaki
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kensei Yamaguchi
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| |
Collapse
|
217
|
Boukhaled GM, Harding S, Brooks DG. Opposing Roles of Type I Interferons in Cancer Immunity. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2021; 16:167-198. [PMID: 33264572 DOI: 10.1146/annurev-pathol-031920-093932] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The immune system is tasked with identifying malignant cells to eliminate or prevent cancer spread. This involves a complex orchestration of many immune cell types that together recognize different aspects of tumor transformation and growth. In response, tumors have developed mechanisms to circumvent immune attack. Type I interferons (IFN-Is) are a class of proinflammatory cytokines produced in response to viruses and other environmental stressors. IFN-Is are also emerging as essential drivers of antitumor immunity, potently stimulating the ability of immune cells to eliminate tumor cells. However, a more complicated role for IFN-Is has arisen, as prolonged stimulation can promote feedback inhibitory mechanisms that contribute to immune exhaustion and other deleterious effects that directly or indirectly permit cancer cells to escape immune clearance. We review the fundamental and opposing functions of IFN-Is that modulate tumor growth and impact immune function and ultimately how these functions can be harnessed for the design of new cancer therapies.
Collapse
Affiliation(s)
- Giselle M Boukhaled
- Princess Margaret Cancer Centre, University Health Network Toronto, Ontario M5G 2M9, Canada; .,Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Shane Harding
- Princess Margaret Cancer Centre, University Health Network Toronto, Ontario M5G 2M9, Canada; .,Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada.,Departments of Medical Biophysics and Radiation Oncology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - David G Brooks
- Princess Margaret Cancer Centre, University Health Network Toronto, Ontario M5G 2M9, Canada; .,Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
218
|
Hepatocellular carcinoma. Nat Rev Dis Primers 2021. [DOI: 10.1038/s41572-020-00240-3 order by 1-- #] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
219
|
Llovet JM, Kelley RK, Villanueva A, Singal AG, Pikarsky E, Roayaie S, Lencioni R, Koike K, Zucman-Rossi J, Finn RS. Hepatocellular carcinoma. Nat Rev Dis Primers 2021; 7:6. [PMID: 33479224 DOI: 10.1038/s41572-020-00240-3] [Citation(s) in RCA: 3672] [Impact Index Per Article: 918.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/08/2020] [Indexed: 02/07/2023]
Abstract
Liver cancer remains a global health challenge, with an estimated incidence of >1 million cases by 2025. Hepatocellular carcinoma (HCC) is the most common form of liver cancer and accounts for ~90% of cases. Infection by hepatitis B virus and hepatitis C virus are the main risk factors for HCC development, although non-alcoholic steatohepatitis associated with metabolic syndrome or diabetes mellitus is becoming a more frequent risk factor in the West. Moreover, non-alcoholic steatohepatitis-associated HCC has a unique molecular pathogenesis. Approximately 25% of all HCCs present with potentially actionable mutations, which are yet to be translated into the clinical practice. Diagnosis based upon non-invasive criteria is currently challenged by the need for molecular information that requires tissue or liquid biopsies. The current major advancements have impacted the management of patients with advanced HCC. Six systemic therapies have been approved based on phase III trials (atezolizumab plus bevacizumab, sorafenib, lenvatinib, regorafenib, cabozantinib and ramucirumab) and three additional therapies have obtained accelerated FDA approval owing to evidence of efficacy. New trials are exploring combination therapies, including checkpoint inhibitors and tyrosine kinase inhibitors or anti-VEGF therapies, or even combinations of two immunotherapy regimens. The outcomes of these trials are expected to change the landscape of HCC management at all evolutionary stages.
Collapse
Affiliation(s)
- Josep M Llovet
- Mount Sinai Liver Cancer Program, Division of Liver Diseases, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Translational Research in Hepatic Oncology, Liver Unit, IDIBAPS, Hospital Clinic, University of Barcelona, Catalonia, Spain. .,Institució Catalana d'Estudis Avançats (ICREA), Barcelona, Catalonia, Spain.
| | - Robin Kate Kelley
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Augusto Villanueva
- Mount Sinai Liver Cancer Program, Division of Liver Diseases, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Amit G Singal
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Eli Pikarsky
- The Lautenberg Center for Immunology and Cancer Research, IMRIC, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Sasan Roayaie
- White Plains Hospital Center for Cancer Care, Montefiore Health, White Plains, NY, USA
| | - Riccardo Lencioni
- Department of Radiology, Pisa University School of Medicine, Pisa, Italy.,Department of Radiology, Miami Cancer Insitute, Miami, FL, USA
| | - Kazuhiko Koike
- The University of Tokyo, Department of Gastroenterology, Tokyo, Japan
| | - Jessica Zucman-Rossi
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, USPC, Université Paris Descartes, Université Paris Diderot, Paris, France.,Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Richard S Finn
- Department of Oncology, Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
220
|
Hepatocellular carcinoma. Nat Rev Dis Primers 2021. [DOI: 10.1038/s41572-020-00240-3 and 1880=1880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
221
|
Hepatocellular carcinoma. Nat Rev Dis Primers 2021. [DOI: 10.1038/s41572-020-00240-3 order by 1-- -] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
222
|
Hepatocellular carcinoma. Nat Rev Dis Primers 2021. [DOI: 10.1038/s41572-020-00240-3 order by 1-- gadu] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
223
|
Tumor-Infiltrating Lymphoid Cells in Colorectal Cancer Patients with Varying Disease Stages and Microsatellite Instability-High/Stable Tumors. Vaccines (Basel) 2021; 9:vaccines9010064. [PMID: 33477864 PMCID: PMC7832866 DOI: 10.3390/vaccines9010064] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 01/10/2021] [Accepted: 01/15/2021] [Indexed: 12/24/2022] Open
Abstract
Immune checkpoint inhibition is an effective anti-cancer therapeutic approach but has shown limited efficacy in treating colorectal cancer (CRC) patients. Importantly, immune constituents of the tumor microenvironment (TME) can influence therapy response and cancer progression. We investigated the expression of immune checkpoints (ICs) on lymphoid populations within the CRC TME and compared with cells from normal colon tissues using samples from 50 patients with varying disease stages. We found that the levels of B cells, T cells, and NK cells were similar, IC-expressing CD4+ and CD4+CD8+ double positive T cells were higher, while CD8+ T cells and CD4−CD8− double negative T cells were significantly lower in CRC tumors. Notably, patients with mismatch-repair deficiency/microsatellite instability-high tumors had higher levels of IC-expressing CD4+ and CD8+ T cells than patients with proficient MMR and microsatellite stable tumors. Lastly, The Cancer Genome Atlas Colon Adenocarcinoma datasets showed associations between low expression of selective genes and poorer progression-free interval. Our findings highlight differential expression of ICs on lymphoid cells in CRC tumors in the era of cancer immunotherapy, which at present is solely approved for anti-PD-1 therapy in patients with dMMR/MSI-H tumors. Further investigations into their functionality have potentials for deciphering resistance mechanisms to IC inhibition.
Collapse
|
224
|
Ioannou N, Hagner PR, Stokes M, Gandhi AK, Apollonio B, Fanous M, Papazoglou D, Sutton LA, Rosenquist R, Amini RM, Chiu H, Lopez-Girona A, Janardhanan P, Awan FT, Jones J, Kay NE, Shanafelt TD, Tallman MS, Stamatopoulos K, Patten PEM, Vardi A, Ramsay AG. Triggering interferon signaling in T cells with avadomide sensitizes CLL to anti-PD-L1/PD-1 immunotherapy. Blood 2021; 137:216-231. [PMID: 33024998 PMCID: PMC7820876 DOI: 10.1182/blood.2020006073] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 09/26/2020] [Indexed: 12/14/2022] Open
Abstract
Cancer treatment has been transformed by checkpoint blockade therapies, with the highest anti-tumor activity of anti-programmed death 1 (PD-1) antibody therapy seen in Hodgkin lymphoma. Disappointingly, response rates have been low in the non-Hodgkin lymphomas, with no activity seen in relapsed/refractory chronic lymphocytic leukemia (CLL) with PD-1 blockade. Thus, identifying more powerful combination therapy is required for these patients. Here, we preclinically demonstrate enhanced anti-CLL activity following combinational therapy with anti-PD-1 or anti-PD-1 ligand (PD-L1) and avadomide, a cereblon E3 ligase modulator (CELMoD). Avadomide induced type I and II interferon (IFN) signaling in patient T cells, triggering a feedforward cascade of reinvigorated T-cell responses. Immune modeling assays demonstrated that avadomide stimulated T-cell activation, chemokine expression, motility and lytic synapses with CLL cells, as well as IFN-inducible feedback inhibition through upregulation of PD-L1. Patient-derived xenograft tumors treated with avadomide were converted to CD8+ T cell-inflamed tumor microenvironments that responded to anti-PD-L1/PD-1-based combination therapy. Notably, clinical analyses showed increased PD-L1 expression on T cells, as well as intratumoral expression of chemokine signaling genes in B-cell malignancy patients receiving avadomide-based therapy. These data illustrate the importance of overcoming a low inflammatory T-cell state to successfully sensitize CLL to checkpoint blockade-based combination therapy.
Collapse
Affiliation(s)
- Nikolaos Ioannou
- School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | | | | | | | - Benedetta Apollonio
- School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Mariam Fanous
- School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Despoina Papazoglou
- School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Lesley-Ann Sutton
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Richard Rosenquist
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Clinical Genetics, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Rose-Marie Amini
- Department of Immunology, Genetics and Pathology, Uppsala University and University Hospital, Uppsala, Sweden
| | | | | | | | - Farrukh T Awan
- Division of Hematology, The Ohio State University Cancer Center, Columbus, OH
| | | | - Neil E Kay
- Division of Hematology, Mayo Clinic, Rochester, MN
| | | | | | - Kostas Stamatopoulos
- Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, Greece
| | - Piers E M Patten
- School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
- Department of Haematology, King's College Hospital NHS Foundation Trust, London, United Kingdom; and
| | - Anna Vardi
- Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, Greece
- Hematology Department and HCT Unit, G. Papanikolaou Hospital, Thessaloniki, Greece
| | - Alan G Ramsay
- School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| |
Collapse
|
225
|
Guruprasad P, Lee YG, Kim KH, Ruella M. The current landscape of single-cell transcriptomics for cancer immunotherapy. J Exp Med 2021; 218:e20201574. [PMID: 33601414 PMCID: PMC7754680 DOI: 10.1084/jem.20201574] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/28/2020] [Accepted: 12/02/2020] [Indexed: 12/28/2022] Open
Abstract
Immunotherapies such as immune checkpoint blockade and adoptive cell transfer have revolutionized cancer treatment, but further progress is hindered by our limited understanding of tumor resistance mechanisms. Emerging technologies now enable the study of tumors at the single-cell level, providing unprecedented high-resolution insights into the genetic makeup of the tumor microenvironment and immune system that bulk genomics cannot fully capture. Here, we highlight the recent key findings of the use of single-cell RNA sequencing to deconvolute heterogeneous tumors and immune populations during immunotherapy. Single-cell RNA sequencing has identified new crucial factors and cellular subpopulations that either promote tumor progression or leave tumors vulnerable to immunotherapy. We anticipate that the strategic use of single-cell analytics will promote the development of the next generation of successful, rationally designed immunotherapeutics.
Collapse
Affiliation(s)
- Puneeth Guruprasad
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
- Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Yong Gu Lee
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
- Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Ki Hyun Kim
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
- Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Marco Ruella
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
- Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
226
|
Xie P, Wang Y, Wei D, Zhang L, Zhang B, Xiao H, Song H, Mao X. Nanoparticle-based drug delivery systems with platinum drugs for overcoming cancer drug resistance. J Mater Chem B 2021; 9:5173-5194. [PMID: 34116565 DOI: 10.1039/d1tb00753j] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Platinum drugs are commonly used in cancer therapy, but their therapeutic outcomes have been significantly compromised by the drug resistance of cancer cells. To this end, intensive efforts have been made to develop nanoparticle-based drug delivery systems for platinum drugs, due to their multifunctionality in delivering drugs, in modulating the tumor microenvironment, and in integrating additional genes, proteins, and small molecules to overcome chemoresistance in cancers. To facilitate the clinical application of these promising nanoparticle-based platinum drug delivery systems, this paper summarizes the common mechanisms for chemoresistance towards platinum drugs, the advantages of nanoparticles in drug delivery, and recent strategies of nanoparticle-based platinum drug delivery. Furthermore, we discuss how to design delivery platforms more effectively to overcome chemoresistance in cancers, thereby improving the efficacy of platinum-based chemotherapy.
Collapse
Affiliation(s)
- Peng Xie
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China. and Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
| | - Yushu Wang
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Dengshuai Wei
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
| | - Lingpu Zhang
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
| | - Bin Zhang
- XJTU-Oxford International Joint Laboratory for Catalysis, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Haihua Xiao
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
| | - Haiqin Song
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai 200025, China.
| | - Xinzhan Mao
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.
| |
Collapse
|
227
|
Shi X, Li CW, Tan LC, Wen SS, Liao T, Zhang Y, Chen TZ, Ma B, Yu PC, Lu ZW, Qu N, Wang Y, Shi RL, Wang YL, Ji QH, Wei WJ. Immune Co-inhibitory Receptors PD-1, CTLA-4, TIM-3, LAG-3, and TIGIT in Medullary Thyroid Cancers: A Large Cohort Study. J Clin Endocrinol Metab 2021; 106:120-132. [PMID: 33000173 DOI: 10.1210/clinem/dgaa701] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Indexed: 12/16/2022]
Abstract
CONTEXT Programmed cell death protein-1 (PD-1), cytotoxic T-lymphocyte antigen 4 (CTLA-4), T-cell immunoglobulin and mucin-domain containing-3 (TIM-3), lymphocyte activation gene-3 (LAG-3), and T-cell immunoglobulin and ITIM domain (TIGIT) are considered major immune co-inhibitory receptors (CIRs) and the most promising immunotherapeutic targets in cancer treatment, but they are largely unexplored in medullary thyroid carcinoma (MTC). OBJECTIVE We aimed to provide the first evidence regarding the expression profiles and clinical significance of CIRs in a large cohort of MTC patients. DESIGN AND PATIENTS In total, 200 MTC patients who received initial surgery in our hospital were included. Immunohistochemistry was performed to evaluate CIR expressions in tissue microarrays (TMAs). Combined with the results of our previous programmed cell death ligand-1 (PD-L1) study, clinicopathologic and prognostic correlations of these proteins were retrospectively analyzed. RESULTS TIM-3, PD-1, CTLA-4, LAG-3, and TIGIT positivity was detected in 96 (48.0%), 27 (13.5%), 25 (12.5%), 6 (3.0%), and 6 (3.0%) patients, respectively, in whom TIM-3, PD-1, and CTLA-4 expressions were positively correlated. Log-rank tests and multivariate Cox analyses both indicated that TIM-3, CTLA-4 expression, and PD-1/PD-L1 coexpression were associated with worse structural recurrence-free survival. In addition, among 20 patients who developed advanced disease during follow-up, 12 (60%) showed TIM-3 positivity, among whom 6 cases also had concurrent moderate to strong PD-1, PD-L1, or CTLA-4 expression. CONCLUSIONS Using the currently largest TMA cohort of this rare cancer, we delineated the CIR expression profiles in MTC, and identified TIM-3, CTLA-4 expression, and PD-1/PD-L1 coexpression as promising biomarkers for tumor recurrence. Furthermore, a subset of advanced MTCs are probably immunogenic, for which single or combined immunotherapy including TIM-3, PD-1, PD-L1, or CTLA-4 blockade may be potential therapeutic approaches in the future.
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Aged, 80 and over
- Antigens, CD/analysis
- Antigens, CD/metabolism
- Biomarkers, Tumor/analysis
- Biomarkers, Tumor/metabolism
- CTLA-4 Antigen/analysis
- CTLA-4 Antigen/metabolism
- Carcinoma, Neuroendocrine/diagnosis
- Carcinoma, Neuroendocrine/metabolism
- Carcinoma, Neuroendocrine/mortality
- Carcinoma, Neuroendocrine/pathology
- Child
- China/epidemiology
- Cohort Studies
- Disease Progression
- Female
- Hepatitis A Virus Cellular Receptor 2/analysis
- Hepatitis A Virus Cellular Receptor 2/metabolism
- Humans
- Immune Checkpoint Proteins/analysis
- Immune Checkpoint Proteins/metabolism
- Immunohistochemistry
- Male
- Middle Aged
- Prognosis
- Programmed Cell Death 1 Receptor/analysis
- Programmed Cell Death 1 Receptor/metabolism
- Receptors, Immunologic/analysis
- Receptors, Immunologic/metabolism
- Retrospective Studies
- Survival Analysis
- Thyroid Neoplasms/diagnosis
- Thyroid Neoplasms/metabolism
- Thyroid Neoplasms/mortality
- Thyroid Neoplasms/pathology
- Tissue Array Analysis
- Young Adult
- Lymphocyte Activation Gene 3 Protein
Collapse
Affiliation(s)
- Xiao Shi
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Cui-Wei Li
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Li-Cheng Tan
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shi-Shuai Wen
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Tian Liao
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yan Zhang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Tong-Zhen Chen
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Ben Ma
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Peng-Cheng Yu
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhong-Wu Lu
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ning Qu
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu Wang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Rong-Liang Shi
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu-Long Wang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qing-Hai Ji
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wen-Jun Wei
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
228
|
Hao X, Falo Iii LD, Chen G, Zhang J, Carey CD, Storkus WJ, Falo LD, You Z. Skin immunization for effective treatment of multifocal melanoma refractory to PD1 blockade and Braf inhibitors. J Immunother Cancer 2021; 9:e001179. [PMID: 33408093 PMCID: PMC7789470 DOI: 10.1136/jitc-2020-001179] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/09/2020] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Despite the remarkable benefits associated with the interventional treatment of melanomas (and other solid cancers) with immune checkpoint and Braf inhibitors (Brafi), most patients ultimately progress on therapy. The presence of multifocal/disseminated disease in patients increases their mortality risk. Hence, the development of novel strategies to effectively treat patients with melanomas that are resistant to anti-PD1 mAb (αPD1) and/or Brafi, particularly those with multifocal/disseminated disease remains a major unmet clinical need. METHODS Mice developing induced/spontaneous BrafV600E/Pten-/- melanomas were treated by cutaneous immunization with a DNA vaccine encoding the melanoma-associated antigen TRP2, with Brafi or αPD1 alone, or with a combination of these treatments. Tumor progression, tumor-infiltration by CD4+ and CD8+ T cells, and the development of TRP2-specific CD8+ T cells were then monitored over time. RESULTS Vaccination led to durable antitumor immunity against PD1/Brafi-resistant melanomas in both single lesion and multifocal disease models, and it sensitized PD1-resistant melanomas to salvage therapy with αPD1. The therapeutic efficacy of the vaccine was associated with host skin-resident cells, the induction of a systemic, broadly reactive IFNγ+CD8+ T cell repertoire, increased frequencies of CD8+ TIL and reduced levels of PD1hi/intCD8+ T cells. Extended survival was associated with improved TIL functionality, exemplified by the presence of enhanced levels of IFNγ+CD8+ TIL and IL2+CD4+ TIL. CONCLUSIONS These data support the use of a novel genetic vaccine for the effective treatment of localized or multifocal melanoma refractory to conventional αPD1-based and/or Brafi-based (immune)therapy.
Collapse
Affiliation(s)
- Xingxing Hao
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Louis D Falo Iii
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Dietrich School of Arts & Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Guo Chen
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jiying Zhang
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Cara D Carey
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Walter J Storkus
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- The University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Louis D Falo
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- The University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, Pennsylvania, USA
- The University of Pittsburgh Clinical and Translational Science Institute, Pittsburgh, Pennsylvania, USA
- The University of Pittsburgh McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania, USA
| | - Zhaoyang You
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
229
|
Sharma S, Kadam P, Singh RP, Davoodi M, St John M, Lee JM. CCL21-DC tumor antigen vaccine augments anti-PD-1 therapy in lung cancer. AIMS MEDICAL SCIENCE 2021. [DOI: 10.3934/medsci.2021022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
<abstract>
<p>Targeting inhibitory immune checkpoint molecules has highlighted the need to find approaches enabling the induction and activation of an immune response against cancer. Therapeutic vaccination, which can induce a specific immune response against tumor antigens, is an important approach to consider. Although this approach has shown low clinical efficacy when combined with other treatment modalities, therapeutic cancer vaccines will have a better outcome when combined with immune checkpoint blockade therapy with potential for cancer free survival. In this review, we will discuss the results of our two recent publications in preclinical lung cancer models. Our studies reveal that anti-PD-1 administered in combination with CCL21-DC tumor antigen therapeutic vaccines eradicate lung cancer. The results of these studies highlight the importance of combination therapy of immune checkpoint blockade and therapeutic cancer vaccines for lung cancer patients.</p>
</abstract>
Collapse
|
230
|
Lee SJ, Wang W, Jin L, Lu X, Gao L, Chen Y, Liu T, Emery D, Vukmanic E, Liu Y, Kaplan HJ, Dean DC. Rod photoreceptor clearance due to misfolded rhodopsin is linked to a DAMP-immune checkpoint switch. J Biol Chem 2021; 296:100102. [PMID: 33214223 PMCID: PMC7949052 DOI: 10.1074/jbc.ra120.016053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 11/13/2020] [Accepted: 11/19/2020] [Indexed: 01/19/2023] Open
Abstract
Chronic endoplasmic reticulum stress resulting from misfolding of the visual pigment rhodopsin (RHO) can lead to loss of rod photoreceptors, which initiates retinitis pigmentosa, characterized initially by diminished nighttime and peripheral vision. Cone photoreceptors depend on rods for glucose transport, which the neurons use for assembly of visual pigment-rich structures; as such, loss of rods also leads to a secondary loss of cone function, diminishing high-resolution color vision utilized for tasks including reading, driving, and facial recognition. If dysfunctional rods could be maintained to continue to serve this secondary cone preservation function, it might benefit patients with retinitis pigmentosa, but the mechanisms by which rods are removed are not fully established. Using pigs expressing mutant RHO, we find that induction of a danger-associated molecular pattern (DAMP) "eat me" signal on the surface of mutant rods is correlated with targeting the live cells for (PrCR) by retinal myeloid cells. Glucocorticoid therapy leads to replacement of this DAMP with a "don't eat me" immune checkpoint on the rod surface and inhibition of PrCR. Surviving rods then continue to promote glucose transport to cones, maintaining their viability.
Collapse
Affiliation(s)
- Sang Joon Lee
- Department of Ophthalmology and Visual Sciences, University of Louisville Health Sciences Center, Louisville, Kentucky, USA; Department of Ophthalmology, Kosin University College of Medicine, Seo-gu, Busan, Korea
| | - Wei Wang
- Department of Ophthalmology and Visual Sciences, University of Louisville Health Sciences Center, Louisville, Kentucky, USA
| | - Lei Jin
- Department of Ophthalmology and Visual Sciences, University of Louisville Health Sciences Center, Louisville, Kentucky, USA; Department of Ophthalmology, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, China
| | - Xiaoqin Lu
- Department of Ophthalmology and Visual Sciences, University of Louisville Health Sciences Center, Louisville, Kentucky, USA; Department of Medicine, University of Louisville Health Sciences Center, Louisville, Kentucky, USA
| | - Lei Gao
- Department of Ophthalmology and Visual Sciences, University of Louisville Health Sciences Center, Louisville, Kentucky, USA; Department of Hematology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yao Chen
- Department of Ophthalmology and Visual Sciences, University of Louisville Health Sciences Center, Louisville, Kentucky, USA; Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
| | - Tingting Liu
- Department of Ophthalmology and Visual Sciences, University of Louisville Health Sciences Center, Louisville, Kentucky, USA; Department of Ophthalmology, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, China
| | - Douglas Emery
- Department of Ophthalmology and Visual Sciences, University of Louisville Health Sciences Center, Louisville, Kentucky, USA; Department of Medicine, University of Louisville Health Sciences Center, Louisville, Kentucky, USA
| | - Eric Vukmanic
- Department of Ophthalmology and Visual Sciences, University of Louisville Health Sciences Center, Louisville, Kentucky, USA; Department of Medicine, University of Louisville Health Sciences Center, Louisville, Kentucky, USA
| | - Yongqing Liu
- Department of Ophthalmology and Visual Sciences, University of Louisville Health Sciences Center, Louisville, Kentucky, USA; Department of Medicine, University of Louisville Health Sciences Center, Louisville, Kentucky, USA
| | - Henry J Kaplan
- Department of Ophthalmology and Visual Sciences, University of Louisville Health Sciences Center, Louisville, Kentucky, USA
| | - Douglas C Dean
- Department of Ophthalmology and Visual Sciences, University of Louisville Health Sciences Center, Louisville, Kentucky, USA; Department of Medicine, University of Louisville Health Sciences Center, Louisville, Kentucky, USA.
| |
Collapse
|
231
|
Wang L, Liu Y, Liu H, Tian H, Wang Y, Zhang G, Lei Y, Xue L, Zheng B, Fan T, Zheng Y, Tan F, Xue Q, Gao S, Li C, He J. The therapeutic significance of the novel photodynamic material TPE-IQ-2O in tumors. Aging (Albany NY) 2020; 13:1383-1409. [PMID: 33472175 PMCID: PMC7835032 DOI: 10.18632/aging.202355] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 10/21/2020] [Indexed: 12/11/2022]
Abstract
Combination therapies based on photodynamic therapy (PDT) have received much attention in various cancers due to their strong therapeutic effects. Here, we aimed to explore the safety and effectiveness of a new mitochondria-targeting photodynamic material, TPE-IQ-2O, in combination therapies (combined with surgery or immunotherapy). The safety and effectiveness of TPE-IQ-2O PDT were verified with cytotoxicity evaluation in vitro and a zebrafish xenograft model in vivo, respectively. The effectiveness of TPE-IQ-2O PDT combined with surgery or immune checkpoint inhibitors (ICIs) was verified in tumor-bearing mice. Small animal in vivo imaging, immunohistochemistry, and flow cytometry were used to determine the underlying mechanism. TPE-IQ-2O PDT can not only reduce tumor recurrence in surgical treatment but also effectively improve the response to ICIs in immunotherapy without obvious toxicity. It was also found to ameliorate the immunosuppressive tumor microenvironment and promote the antitumor immunity induced by ICIs by increasing CD8+ tumor-infiltrating lymphocyte accumulation. Thus, TPE-IQ-2O PDT is a safe and effective antitumor therapy that can be combined with surgery or immunotherapy.
Collapse
Affiliation(s)
- Liyu Wang
- Department of Thoracic Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yu Liu
- Department of Thoracic Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Hengchang Liu
- Department of Colorectal Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - He Tian
- Department of Thoracic Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yalong Wang
- Department of Thoracic Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Guochao Zhang
- Department of Thoracic Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yuanyuan Lei
- Department of Thoracic Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Liyan Xue
- Department of Pathology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Bo Zheng
- Department of Pathology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Tao Fan
- Department of Thoracic Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yujia Zheng
- Department of Thoracic Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Fengwei Tan
- Department of Thoracic Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Qi Xue
- Department of Thoracic Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Shugeng Gao
- Department of Thoracic Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Chunxiang Li
- Department of Thoracic Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
232
|
Cancer Immunotherapy and Application of Nanoparticles in Cancers Immunotherapy as the Delivery of Immunotherapeutic Agents and as the Immunomodulators. Cancers (Basel) 2020; 12:cancers12123773. [PMID: 33333816 PMCID: PMC7765190 DOI: 10.3390/cancers12123773] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/09/2020] [Accepted: 12/10/2020] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Cancer becomes one of the major public health problems globally and the burden is expected to be increasing. Currently, both the medical and research communities have attempted an approach to nonconventional cancer therapies that can limit damage or loss of healthy tissues and be able to fully eradicate the cancer cells. In the last few decades, cancer immunotherapy becomes an important tactic for cancer treatment. Immunotherapy of cancer must activate the host’s anti-tumor response by enhancing the innate immune system and the effector cell number, while, minimizing the host’s suppressor mechanisms. However, many immunotherapies are still limited by poor therapeutic targeting and unwanted side effects. Hence, a deeper understanding of tumor immunology and antitumor immune responses is essential for further improvement of cancer immunotherapy. In addition, effective delivery systems are required to deliver immunotherapeutic agents to the site of interest (such as: to Tumor microenvironments, to Antigen-Presenting Cells, and to the other immune systems) to enhance their efficacy by minimizing off-targeted and unwanted cytotoxicity. Abstract In the last few decades, cancer immunotherapy becomes an important tactic for cancer treatment. However, some immunotherapy shows certain limitations including poor therapeutic targeting and unwanted side effects that hinder its use in clinics. Recently, several researchers are exploring an alternative methodology to overcome the above limitations. One of the emerging tracks in this field area is nano-immunotherapy which has gone through rapid progress and revealed considerable potentials to solve limitations related to immunotherapy. Targeted and stimuli-sensitive biocompatible nanoparticles (NPs) can be synthesized to deliver immunotherapeutic agents in their native conformations to the site of interest to enhance their antitumor activity and to enhance the survival rate of cancer patients. In this review, we have discussed cancer immunotherapy and the application of NPs in cancer immunotherapy, as a carrier of immunotherapeutic agents and as a direct immunomodulator.
Collapse
|
233
|
Filippi L, Nervi C, Proietti I, Pirisino R, Potenza C, Martelli O, Equitani F, Bagni O. Molecular imaging in immuno-oncology: current status and translational perspectives. Expert Rev Mol Diagn 2020; 20:1199-1211. [PMID: 33215963 DOI: 10.1080/14737159.2020.1854090] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Only 20-40% of patients respond to therapy with immune checkpoint inhibitors (ICIs). Therefore, the early identification of subjects that can benefit from such therapeutic regimen is mandatory. Areas covered: The immunobiological mechanisms of ICIs are briefly illustrated. Furthermore, the limitations of traditional radiological approaches are covered. Then, the pros and cons of molecular imaging through positron emission computed tomography (PET/CT) are reviewed, with a particular focus on 18f-fluorodeoxyglucose (18F-FDG) and PET-derived metabolic parameters. Lastly, translational perspective of radiopharmaceuticals others than 18F-FDG such as 89zirconium (89Zr) or fluorine-18 (18F) labeled monoclonal antibodies (e.g.89Zr-atezolizumab, 89Zr-nivolumab) binding to specific biomarkers are discussed. Expert opinion: Molecular imaging presents a prominent role for the management of oncological patients treated with ICIs. Preliminary clinical data indicate that PET/CT with 18F-FDG is useful for assessing the response to treatment and for the imaging of immune-related adverse effects. Nevertheless, the methodological approach (iPERCIST, PERCIMT, or others) to be used for an optimal diagnostic accuracy and patients' evaluation is still a debated issue. PET/CT with radioligands directed toward ICIs biomarkers, although is still in a translational phase, holds the promise of accurately predicting the response to treatment and revealing the acquired resistance to immunotherapy.
Collapse
Affiliation(s)
- Luca Filippi
- Department of Nuclear Medicine, Santa Maria Goretti Hospital, AUSL , Latina, Italy
| | - Clara Nervi
- Department of Medical and Surgical Sciences and Biotechnology, University of Rome "La Sapienza" , Latina, Italy
| | - Ilaria Proietti
- Dermatology Unit Daniele Innocenzi, A. Fiorini Hospital, Polo Pontino , Terracina, Italy
| | - Riccardo Pirisino
- Department of Nuclear Medicine, Santa Maria Goretti Hospital, AUSL , Latina, Italy
| | - Concetta Potenza
- Dermatology Unit Daniele Innocenzi, A. Fiorini Hospital, Polo Pontino , Terracina, Italy
| | | | - Francesco Equitani
- Department of Transfusion Medicine, Santa Maria Goretti Hospital, AUSL , Latina, Italy
| | - Oreste Bagni
- Department of Nuclear Medicine, Santa Maria Goretti Hospital, AUSL , Latina, Italy
| |
Collapse
|
234
|
Cancer and Immune Checkpoint Inhibitor Treatment in the Era of SARS-CoV-2 Infection. Cancers (Basel) 2020; 12:cancers12113383. [PMID: 33207589 PMCID: PMC7698088 DOI: 10.3390/cancers12113383] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/13/2020] [Accepted: 11/13/2020] [Indexed: 12/17/2022] Open
Abstract
Simple Summary The introduction of immune checkpoint inhibitors (ICI) in 2011 revolutionized the management of many solid cancers and hematological malignancies. However, there are concerns regarding the use of ICI in the era of COVID-19. We present currently available information on the pros and cons of using ICI in cancer patients with respect to the risk of acquiring an infection by SARS-CoV2 and mortality from COVID-19. By means of the present paper, clinicians and researchers may update their knowledge on a highly topical clinical question—is the use of ICI in cancer patients with SARS-CoV2 infection harmful with respect to COVID-19 outcome? Abstract Whether cancer patients receiving immune checkpoint inhibitors (ICI) are at an increased risk of severe infection and mortality during the corona pandemic is a hotly debated topic that will continue to evolve. Here, we summarize and discuss current studies regarding COVID-19 and anti-cancer treatment with an emphasis on ICI. Importantly, several lines of evidence suggest that patients currently treated with ICI do not display an increased vulnerability to infection with SARS-CoV-2. Data regarding morbidity and mortality associated with COVID-19 in cancer patients receiving ICI are less clear and often conflicting. Although mostly based on experimental data, it is possible that ICI can promote the exacerbated immune response associated with adverse outcome in COVID-19 patients. On the other hand, mounting evidence suggests that ICI might even be useful in the treatment of viral infections by preventing or ameliorating T cell exhaustion. In this context, the right timing of treatment might be essential. Nevertheless, some cancer patients treated with ICI experience autoimmune-related side effects that require the use of immunosuppressive therapies, which in turn may promote a severe course of infection with SARS-CoV-2. Although there is clear evidence that withholding ICI will have more serious consequences, further studies are urgently needed in to better evaluate the effects of ICI in patients with COVID-19 and the use of ICI during the corona pandemic in general.
Collapse
|
235
|
Hwang JK, Hong J, Yun CO. Oncolytic Viruses and Immune Checkpoint Inhibitors: Preclinical Developments to Clinical Trials. Int J Mol Sci 2020; 21:E8627. [PMID: 33207653 PMCID: PMC7697902 DOI: 10.3390/ijms21228627] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 10/30/2020] [Accepted: 11/06/2020] [Indexed: 12/12/2022] Open
Abstract
Immuno-oncology (IO) has been an active area of oncology research. Following US FDA approval of the first immune checkpoint inhibitor (ICI), ipilimumab (human IgG1 k anti-CTLA-4 monoclonal antibody), in 2011, and of the first oncolytic virus, Imlygic (talimogene laherparepvec), in 2015, there has been renewed interest in IO. In the past decade, ICIs have changed the treatment paradigm for many cancers by enabling better therapeutic control, resuming immune surveillance, suppressing tumor immunosuppression, and restoring antitumor immune function. However, ICI therapies are effective only in a small subset of patients and show limited therapeutic potential due to their inability to demonstrate efficacy in 'cold' or unresponsive tumor microenvironments (TMEs). Relatedly, oncolytic viruses (OVs) have been shown to induce antitumor immune responses, augment the efficacy of existing cancer treatments, and reform unresponsive TME to turn 'cold' tumors 'hot,' increasing their susceptibility to checkpoint blockade immunotherapies. For this reason, OVs serve as ideal complements to ICIs, and multiple preclinical studies and clinical trials are demonstrating their combined therapeutic efficacy. This review will discuss the merits and limitations of OVs and ICIs as monotherapy then progress onto the preclinical rationale and the results of clinical trials of key combination therapies.
Collapse
Affiliation(s)
- June Kyu Hwang
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Korea; (J.K.H.); (J.H.)
| | - JinWoo Hong
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Korea; (J.K.H.); (J.H.)
- GeneMedicine Co., Ltd., 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Korea
| | - Chae-Ok Yun
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Korea; (J.K.H.); (J.H.)
- GeneMedicine Co., Ltd., 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Korea
- Institute of Nano Science and Technology, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Korea
| |
Collapse
|
236
|
Huang X, Tang T, Zhang G, Hong Z, Xu J, Yadav DK, Bai X, Liang T. Genomic investigation of co-targeting tumor immune microenvironment and immune checkpoints in pan-cancer immunotherapy. NPJ Precis Oncol 2020; 4:29. [PMID: 33299118 PMCID: PMC7666137 DOI: 10.1038/s41698-020-00136-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 10/21/2020] [Indexed: 12/12/2022] Open
Abstract
Drugs that target immune checkpoints (ICPs) have become the most popular weapons in cancer immunotherapy; however, they are only beneficial for a small fraction of patients. Accumulating evidence suggests that the tumor immune microenvironment (TIME) plays a critical role in anti-cancer immunity. This study aimed to assess the potential merits and feasibility of combinational targeting ICPs and TIME in cancer immunotherapy. A total of 31 cancer type-specific datasets in TCGA were individually collected by the publicly available web servers for multiple bioinformatic analyses of ICPs and TIME factors. GEPIA was used to calculate the prognostic indexes, STRING was used to construct protein-protein interactions, cBioPortal was used for visualization and comparison of genetic alterations, and TISIDB was used to explore the correlation to tumor-infiltrating lymphocytes (TILs). Intriguingly, TIME factors were identified to have more global coverage and prognostic significance across multiple cancer types compared with ICPs, thus offering more general targetability in clinical therapy. Moreover, TIME factors showed interactive potential with ICPs, and genomic alteration of TIME factors coupled with that of ICPs, at least in pancreatic cancer. Furthermore, TIME factors were found to be significantly associated with TILs, including but not limited to pancreatic cancer. Finally, the clinical significance and translational potential of further combination therapies that incorporate both ICP inhibitors and TIME factor-targeted treatments were discussed. Together, TIME factors are promising immunotherapeutic targets, and a combination strategy of TIME factors-targeted therapies with ICP inhibitors may benefit more cancer patients in the future.
Collapse
Affiliation(s)
- Xing Huang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China.
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China.
- Innovation Center for the Study of Pancreatic Diseases, Zhejiang Province, Hangzhou, 310003, Zhejiang, China.
| | - Tianyu Tang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
- Innovation Center for the Study of Pancreatic Diseases, Zhejiang Province, Hangzhou, 310003, Zhejiang, China
| | - Gang Zhang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
- Innovation Center for the Study of Pancreatic Diseases, Zhejiang Province, Hangzhou, 310003, Zhejiang, China
| | - Zhengtao Hong
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
- Innovation Center for the Study of Pancreatic Diseases, Zhejiang Province, Hangzhou, 310003, Zhejiang, China
| | - Jian Xu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
- Innovation Center for the Study of Pancreatic Diseases, Zhejiang Province, Hangzhou, 310003, Zhejiang, China
| | - Dipesh Kumar Yadav
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
- Innovation Center for the Study of Pancreatic Diseases, Zhejiang Province, Hangzhou, 310003, Zhejiang, China
| | - Xueli Bai
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China.
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China.
- Innovation Center for the Study of Pancreatic Diseases, Zhejiang Province, Hangzhou, 310003, Zhejiang, China.
| | - Tingbo Liang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China.
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China.
- Innovation Center for the Study of Pancreatic Diseases, Zhejiang Province, Hangzhou, 310003, Zhejiang, China.
| |
Collapse
|
237
|
Papavassiliou AG, Musti AM. The Multifaceted Output of c-Jun Biological Activity: Focus at the Junction of CD8 T Cell Activation and Exhaustion. Cells 2020; 9:2470. [PMID: 33202877 PMCID: PMC7697663 DOI: 10.3390/cells9112470] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/07/2020] [Accepted: 11/11/2020] [Indexed: 12/19/2022] Open
Abstract
c-Jun is a major component of the dimeric transcription factor activator protein-1 (AP-1), a paradigm for transcriptional response to extracellular signaling, whose components are basic-Leucine Zipper (bZIP) transcription factors of the Jun, Fos, activating transcription factor (ATF), ATF-like (BATF) and Jun dimerization protein 2 (JDP2) gene families. Extracellular signals regulate c-Jun/AP-1 activity at multiple levels, including transcriptional and posttranscriptional regulation of c-Jun expression and transactivity, in turn, establishing the magnitude and the duration of c-Jun/AP-1 activation. Another important level of c-Jun/AP-1 regulation is due to the capability of Jun family members to bind DNA as a heterodimer with every other member of the AP-1 family, and to interact with other classes of transcription factors, thereby acquiring the potential to integrate diverse extrinsic and intrinsic signals into combinatorial regulation of gene expression. Here, we review how these features of c-Jun/AP-1 regulation underlie the multifaceted output of c-Jun biological activity, eliciting quite distinct cellular responses, such as neoplastic transformation, differentiation and apoptosis, in different cell types. In particular, we focus on the current understanding of the role of c-Jun/AP-1 in the response of CD8 T cells to acute infection and cancer. We highlight the transcriptional and epigenetic regulatory mechanisms through which c-Jun/AP-1 participates in the productive immune response of CD8 T cells, and how its downregulation may contribute to the dysfunctional state of tumor infiltrating CD8 T cells. Additionally, we discuss recent insights pointing at c-Jun as a suitable target for immunotherapy-based combination approaches to reinvigorate anti-tumor immune functions.
Collapse
Affiliation(s)
- Athanasios G. Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Anna Maria Musti
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| |
Collapse
|
238
|
Hack SP, Zhu AX, Wang Y. Augmenting Anticancer Immunity Through Combined Targeting of Angiogenic and PD-1/PD-L1 Pathways: Challenges and Opportunities. Front Immunol 2020; 11:598877. [PMID: 33250900 PMCID: PMC7674951 DOI: 10.3389/fimmu.2020.598877] [Citation(s) in RCA: 170] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 10/08/2020] [Indexed: 12/11/2022] Open
Abstract
Cancer immunotherapy (CIT) with antibodies targeting the programmed cell death 1 protein (PD-1)/programmed cell death 1 ligand 1 (PD-L1) axis have changed the standard of care in multiple cancers. However, durable antitumor responses have been observed in only a minority of patients, indicating the presence of other inhibitory mechanisms that act to restrain anticancer immunity. Therefore, new therapeutic strategies targeted against other immune suppressive mechanisms are needed to enhance anticancer immunity and maximize the clinical benefit of CIT in patients who are resistant to immune checkpoint inhibition. Preclinical and clinical studies have identified abnormalities in the tumor microenvironment (TME) that can negatively impact the efficacy of PD-1/PD-L1 blockade. Angiogenic factors such as vascular endothelial growth factor (VEGF) drive immunosuppression in the TME by inducing vascular abnormalities, suppressing antigen presentation and immune effector cells, or augmenting the immune suppressive activity of regulatory T cells, myeloid-derived suppressor cells, and tumor-associated macrophages. In turn, immunosuppressive cells can drive angiogenesis, thereby creating a vicious cycle of suppressed antitumor immunity. VEGF-mediated immune suppression in the TME and its negative impact on the efficacy of CIT provide a therapeutic rationale to combine PD-1/PD-L1 antibodies with anti-VEGF drugs in order to normalize the TME. A multitude of clinical trials have been initiated to evaluate combinations of a PD-1/PD-L1 antibody with an anti-VEGF in a variety of cancers. Recently, the positive results from five Phase III studies in non-small cell lung cancer (adenocarcinoma), renal cell carcinoma, and hepatocellular carcinoma have shown that combinations of PD-1/PD-L1 antibodies and anti-VEGF agents significantly improved clinical outcomes compared with respective standards of care. Such combinations have been approved by health authorities and are now standard treatment options for renal cell carcinoma, non-small cell lung cancer, and hepatocellular carcinoma. A plethora of other randomized studies of similar combinations are currently ongoing. Here, we discuss the principle mechanisms of VEGF-mediated immunosuppression studied in preclinical models or as part of translational clinical studies. We also discuss data from recently reported randomized clinical trials. Finally, we discuss how these concepts and approaches can be further incorporated into clinical practice to improve immunotherapy outcomes for patients with cancer.
Collapse
Affiliation(s)
- Stephen P. Hack
- Product Development (Oncology), Genentech, Inc., South San Francisco, CA, United States
| | - Andrew X. Zhu
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, United States
- Jiahui International Cancer Center, Jiahui Health, Shanghai, China
| | - Yulei Wang
- Product Development (Oncology), Genentech, Inc., South San Francisco, CA, United States
| |
Collapse
|
239
|
Cichocki F, Bjordahl R, Gaidarova S, Mahmood S, Abujarour R, Wang H, Tuininga K, Felices M, Davis ZB, Bendzick L, Clarke R, Stokely L, Rogers P, Ge M, Robinson M, Rezner B, Robbins DL, Lee TT, Kaufman DS, Blazar BR, Valamehr B, Miller JS. iPSC-derived NK cells maintain high cytotoxicity and enhance in vivo tumor control in concert with T cells and anti-PD-1 therapy. Sci Transl Med 2020; 12:eaaz5618. [PMID: 33148626 PMCID: PMC8861807 DOI: 10.1126/scitranslmed.aaz5618] [Citation(s) in RCA: 172] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 06/05/2020] [Accepted: 09/22/2020] [Indexed: 08/10/2023]
Abstract
The development of immunotherapeutic monoclonal antibodies targeting checkpoint inhibitory receptors, such as programmed cell death 1 (PD-1), or their ligands, such as PD-L1, has transformed the oncology landscape. However, durable tumor regression is limited to a minority of patients. Therefore, combining immunotherapies with those targeting checkpoint inhibitory receptors is a promising strategy to bolster antitumor responses and improve response rates. Natural killer (NK) cells have the potential to augment checkpoint inhibition therapies, such as PD-L1/PD-1 blockade, because NK cells mediate both direct tumor lysis and T cell activation and recruitment. However, sourcing donor-derived NK cells for adoptive cell therapy has been limited by both cell number and quality. Thus, we developed a robust and efficient manufacturing system for the differentiation and expansion of high-quality NK cells derived from induced pluripotent stem cells (iPSCs). iPSC-derived NK (iNK) cells produced inflammatory cytokines and exerted strong cytotoxicity against an array of hematologic and solid tumors. Furthermore, we showed that iNK cells recruit T cells and cooperate with T cells and anti-PD-1 antibody, further enhancing inflammatory cytokine production and tumor lysis. Because the iNK cell derivation process uses a renewable starting material and enables the manufacturing of large numbers of doses from a single manufacture, iNK cells represent an "off-the-shelf" source of cells for immunotherapy with the capacity to target tumors and engage the adaptive arm of the immune system to make a "cold" tumor "hot" by promoting the influx of activated T cells to augment checkpoint inhibitor therapies.
Collapse
Affiliation(s)
- Frank Cichocki
- Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | | | | - Hongbo Wang
- Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Katie Tuininga
- Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Martin Felices
- Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Zachary B Davis
- Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Laura Bendzick
- Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | | - Moyar Ge
- Fate Therapeutics, San Diego, CA 92121, USA
| | | | | | | | - Tom T Lee
- Fate Therapeutics, San Diego, CA 92121, USA
| | - Dan S Kaufman
- Department of Medicine, Division of Regenerative Medicine, Moores Cancer Center, and Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Bruce R Blazar
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
| | | | - Jeffrey S Miller
- Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
240
|
Kumbhari A, Egelston CA, Lee PP, Kim PS. Mature Dendritic Cells May Promote High-Avidity Tuning of Vaccine T Cell Responses. Front Immunol 2020; 11:584680. [PMID: 33193401 PMCID: PMC7662095 DOI: 10.3389/fimmu.2020.584680] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 10/05/2020] [Indexed: 12/12/2022] Open
Abstract
Therapeutic vaccines can elicit tumor-specific cytotoxic T lymphocytes (CTLs), but durable reductions in tumor burden require vaccines that stimulate high-avidity CTLs. Recent advances in immunotherapy responses have led to renewed interest in vaccine approaches, including dendritic cell vaccine strategies. However, dendritic cell requirements for vaccines that generate potent anti-tumor T-cell responses are unclear. Here we use mathematical modeling to show that, counterintuitively, increasing levels of immature dendritic cells may lead to selective expansion of high-avidity CTLs. This finding is in contrast with traditional dendritic cell vaccine approaches that have sought to harness ex vivo generated mature dendritic cells. We show that the injection of vaccine antigens in the context of increased numbers of immature dendritic cells results in a decreased overall peptide:MHC complex load that favors high-avidity CTL activation and expansion. Overall, our results provide a firm basis for further development of this approach, both alone and in combination with other immunotherapies such as checkpoint blockade.
Collapse
Affiliation(s)
- Adarsh Kumbhari
- School of Mathematics and Statistics, University of Sydney, Sydney, NSW, Australia
| | - Colt A. Egelston
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA, United States
| | - Peter P. Lee
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA, United States
| | - Peter S. Kim
- School of Mathematics and Statistics, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
241
|
Aguadé-Gorgorió G, Solé R. Tumour neoantigen heterogeneity thresholds provide a time window for combination immunotherapy. J R Soc Interface 2020; 17:20200736. [PMID: 33109023 DOI: 10.1098/rsif.2020.0736] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Following the advent of cancer immunotherapy, increasing insight has been gained on the role of mutational load and neoantigens as key ingredients in T cell recognition of malignancies. However, not all highly mutational tumours react to immune therapies, and initial success is often followed by eventual relapse. Heterogeneity in the neoantigen landscape of a tumour might be key in the failure of immune surveillance. In this work, we present a mathematical framework to describe how neoantigen distributions shape the immune response. The model predicts the existence of an antigen diversity threshold level beyond which T cells fail at controlling heterogeneous tumours. Incorporating this diversity marker adds predictive value to antigen load for two cohorts of anti-CTLA-4 treated melanoma patients. Furthermore, our analytical approach indicates rapid increases in epitope heterogeneity in early malignancy growth following immune escape. We propose a combination therapy scheme that takes advantage of preexisting resistance to a targeted agent. The model indicates that the selective sweep for a resistant subclone reduces neoantigen heterogeneity, and we postulate the existence of a time window before tumour relapse where checkpoint blockade immunotherapy can become more effective.
Collapse
Affiliation(s)
- Guim Aguadé-Gorgorió
- ICREA-Complex Systems Lab, Universitat Pompeu Fabra, 08003 Barcelona, Spain.,Institut de Biologia Evolutiva (CSIC-UPF), Psg Maritim Barceloneta, 37, 08003 Barcelona, Spain
| | - Ricard Solé
- ICREA-Complex Systems Lab, Universitat Pompeu Fabra, 08003 Barcelona, Spain.,Institut de Biologia Evolutiva (CSIC-UPF), Psg Maritim Barceloneta, 37, 08003 Barcelona, Spain.,Santa Fe Institute, 399 Hyde Park Road, Santa Fe NM 87501, USA
| |
Collapse
|
242
|
Rundqvist H, Veliça P, Barbieri L, Gameiro PA, Bargiela D, Gojkovic M, Mijwel S, Reitzner SM, Wulliman D, Ahlstedt E, Ule J, Östman A, Johnson RS. Cytotoxic T-cells mediate exercise-induced reductions in tumor growth. eLife 2020; 9:e59996. [PMID: 33095157 PMCID: PMC7584454 DOI: 10.7554/elife.59996] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 09/29/2020] [Indexed: 12/13/2022] Open
Abstract
Exercise has a wide range of systemic effects. In animal models, repeated exertion reduces malignant tumor progression, and clinically, exercise can improve outcome for cancer patients. The etiology of the effects of exercise on tumor progression are unclear, as are the cellular actors involved. We show here that in mice, exercise-induced reduction in tumor growth is dependent on CD8+ T cells, and that metabolites produced in skeletal muscle and excreted into plasma at high levels during exertion in both mice and humans enhance the effector profile of CD8+ T-cells. We found that activated murine CD8+ T cells alter their central carbon metabolism in response to exertion in vivo, and that immune cells from trained mice are more potent antitumor effector cells when transferred into tumor-bearing untrained animals. These data demonstrate that CD8+ T cells are metabolically altered by exercise in a manner that acts to improve their antitumoral efficacy.
Collapse
Affiliation(s)
- Helene Rundqvist
- Department of Cell and Molecular Biology, Karolinska InstitutetStockholmSweden
- Department of Laboratory Medicine, Karolinska InstitutetStockholmSweden
| | - Pedro Veliça
- Department of Cell and Molecular Biology, Karolinska InstitutetStockholmSweden
| | - Laura Barbieri
- Department of Cell and Molecular Biology, Karolinska InstitutetStockholmSweden
- Department of Surgery, Oncology, and Gastroenterology, University of PadovaPaduaItaly
| | | | - David Bargiela
- Department of Cell and Molecular Biology, Karolinska InstitutetStockholmSweden
- Department of Physiology, Development, and Neuroscience, University of CambridgeCambridgeUnited Kingdom
| | - Milos Gojkovic
- Department of Cell and Molecular Biology, Karolinska InstitutetStockholmSweden
| | - Sara Mijwel
- Department of Physiology and Pharmacology, Karolinska InstitutetStockholmSweden
| | | | - David Wulliman
- Department of Cell and Molecular Biology, Karolinska InstitutetStockholmSweden
| | - Emil Ahlstedt
- Department of Laboratory Medicine, Karolinska InstitutetStockholmSweden
| | - Jernej Ule
- The Francis Crick InstituteLondonUnited Kingdom
| | - Arne Östman
- Department of Oncology-Pathology, Karolinska InstitutetStockholmSweden
| | - Randall S Johnson
- Department of Cell and Molecular Biology, Karolinska InstitutetStockholmSweden
- Department of Physiology, Development, and Neuroscience, University of CambridgeCambridgeUnited Kingdom
| |
Collapse
|
243
|
Li L, Deng L, Meng X, Gu C, Meng L, Li K, Zhang X, Meng Y, Xu W, Zhao L, Chen J, Zhu Z, Huang H. Tumor-targeting anti-EGFR x anti-PD1 bispecific antibody inhibits EGFR-overexpressing tumor growth by combining EGFR blockade and immune activation with direct tumor cell killing. Transl Oncol 2020; 14:100916. [PMID: 33129108 PMCID: PMC7585148 DOI: 10.1016/j.tranon.2020.100916] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 09/19/2020] [Accepted: 10/12/2020] [Indexed: 12/13/2022] Open
Abstract
The anti-PD1 x anti-EGFR bispecific antibody (BsAb) exhibited all in-vitro bioactivities comparable to that of the parental mAbs and showed anti-tumor efficacies of each of the two arms on par with the mAbs in-vivo. The anti-PD1 x anti-EGFR bispecific antibody (BsAb) retained full ADCC towards cancer cells but not to T cells. Thus the BsAb is capable of killing tumor cells via ADCC while sparing T cells for T cell-induced anti-tumor immunity. The anti-PD1 x anti-EGFR bispecific antibody (BsAb) exhibited significantly stronger tumor cell killing effects in the presence of PBMC relative to that of combination of cetuximab with an anti-PD1 mAb, 609A.
We developed a strategy to combine conventional targeted therapy with immune checkpoint blockade using a tumor-targeting bispecific antibody (BsAb) to treat solid tumors. The BsAb was designed to simultaneously engage a tumor-associated antigen, epidermal growth factor receptor (EGFR), and programed cell death protein 1 (PD1). In addition to its direct anti-tumor activity via EGFR inhibition, the BsAb mediated efficient antibody-dependent cellular cytotoxicity (ADCC) and activated T cell antitumor im munity through blockade of PD1 from interacting with its counterpart, programed cell death ligand 1 (PDL1). Further, the BsAb exhibited a potent direct tumor cell killing activity in the presence of PBMC, most likely, via activating and, at the same time, physically engaging T cells with tumor cells. Taken together, we here illustrate a new strategy in the design and production of novel BsAbs with enhanced therapeutic efficacy through both direct tumor growth inhibition and T cell activation via tumor-targeted immune checkpoint blockade.
Collapse
Affiliation(s)
- Li Li
- Sunshine Guojian Pharmaceutical (Shanghai) Co. Ltd. a 3SBio Inc. Company, 399 Libing Road, Shanghai 201203, China
| | - Lan Deng
- Sunshine Guojian Pharmaceutical (Shanghai) Co. Ltd. a 3SBio Inc. Company, 399 Libing Road, Shanghai 201203, China
| | - Xiaoqing Meng
- Sunshine Guojian Pharmaceutical (Shanghai) Co. Ltd. a 3SBio Inc. Company, 399 Libing Road, Shanghai 201203, China
| | - Changling Gu
- Sunshine Guojian Pharmaceutical (Shanghai) Co. Ltd. a 3SBio Inc. Company, 399 Libing Road, Shanghai 201203, China
| | - Li Meng
- Sunshine Guojian Pharmaceutical (Shanghai) Co. Ltd. a 3SBio Inc. Company, 399 Libing Road, Shanghai 201203, China
| | - Kai Li
- Sunshine Guojian Pharmaceutical (Shanghai) Co. Ltd. a 3SBio Inc. Company, 399 Libing Road, Shanghai 201203, China
| | - Xuesai Zhang
- Sunshine Guojian Pharmaceutical (Shanghai) Co. Ltd. a 3SBio Inc. Company, 399 Libing Road, Shanghai 201203, China
| | - Yun Meng
- Sunshine Guojian Pharmaceutical (Shanghai) Co. Ltd. a 3SBio Inc. Company, 399 Libing Road, Shanghai 201203, China
| | - Wei Xu
- Sunshine Guojian Pharmaceutical (Shanghai) Co. Ltd. a 3SBio Inc. Company, 399 Libing Road, Shanghai 201203, China
| | - Le Zhao
- Sunshine Guojian Pharmaceutical (Shanghai) Co. Ltd. a 3SBio Inc. Company, 399 Libing Road, Shanghai 201203, China
| | - Jianhe Chen
- Sunshine Guojian Pharmaceutical (Shanghai) Co. Ltd. a 3SBio Inc. Company, 399 Libing Road, Shanghai 201203, China
| | - Zhenping Zhu
- Sunshine Guojian Pharmaceutical (Shanghai) Co. Ltd. a 3SBio Inc. Company, 399 Libing Road, Shanghai 201203, China.
| | - Haomin Huang
- Sunshine Guojian Pharmaceutical (Shanghai) Co. Ltd. a 3SBio Inc. Company, 399 Libing Road, Shanghai 201203, China.
| |
Collapse
|
244
|
Kuryk L, Bertinato L, Staniszewska M, Pancer K, Wieczorek M, Salmaso S, Caliceti P, Garofalo M. From Conventional Therapies to Immunotherapy: Melanoma Treatment in Review. Cancers (Basel) 2020; 12:cancers12103057. [PMID: 33092131 PMCID: PMC7589099 DOI: 10.3390/cancers12103057] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/16/2020] [Accepted: 10/18/2020] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Here, we review the current state of knowledge in the field of cancer immunotherapy, focusing on the scientific rationale for the use of oncolytic viruses, checkpoint inhibitors and their combination to combat melanomas. Attention is also given to the immunological aspects of cancer therapy and the shift from conventional therapy towards immunotherapy. This review brings together information on how immunotherapy can be applied to support other cancer therapies in order to maximize the efficacy of melanoma treatment and improve clinical outcomes. Abstract In this review, we discuss the use of oncolytic viruses and checkpoint inhibitors in cancer immunotherapy in melanoma, with a particular focus on combinatory therapies. Oncolytic viruses are promising and novel anti-cancer agents, currently under investigation in many clinical trials both as monotherapy and in combination with other therapeutics. They have shown the ability to exhibit synergistic anticancer activity with checkpoint inhibitors, chemotherapy, radiotherapy. A coupling between oncolytic viruses and checkpoint inhibitors is a well-accepted strategy for future cancer therapies. However, eradicating advanced cancers and tailoring the immune response for complete tumor clearance is an ongoing problem. Despite current advances in cancer research, monotherapy has shown limited efficacy against solid tumors. Therefore, current improvements in virus targeting, genetic modification, enhanced immunogenicity, improved oncolytic properties and combination strategies have a potential to widen the applications of immuno-oncology (IO) in cancer treatment. Here, we summarize the strategy of combinatory therapy with an oncolytic vector to combat melanoma and highlight the need to optimize current practices and improve clinical outcomes.
Collapse
Affiliation(s)
- Lukasz Kuryk
- Department of Virology, National Institute of Public Health-National Institute of Hygiene, Chocimska 24, 00-791 Warsaw, Poland; (K.P.); (M.W.)
- Clinical Science, Targovax Oy, Saukonpaadenranta 2, 00180 Helsinki, Finland
- Correspondence: (L.K.); (M.G.)
| | - Laura Bertinato
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy; (L.B.); (S.S.); (P.C.)
| | - Monika Staniszewska
- Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland;
- Centre for Advanced Materials and Technologies, Warsaw University of Technology, Poleczki 19, 02-822 Warsaw, Poland
| | - Katarzyna Pancer
- Department of Virology, National Institute of Public Health-National Institute of Hygiene, Chocimska 24, 00-791 Warsaw, Poland; (K.P.); (M.W.)
| | - Magdalena Wieczorek
- Department of Virology, National Institute of Public Health-National Institute of Hygiene, Chocimska 24, 00-791 Warsaw, Poland; (K.P.); (M.W.)
| | - Stefano Salmaso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy; (L.B.); (S.S.); (P.C.)
| | - Paolo Caliceti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy; (L.B.); (S.S.); (P.C.)
| | - Mariangela Garofalo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy; (L.B.); (S.S.); (P.C.)
- Correspondence: (L.K.); (M.G.)
| |
Collapse
|
245
|
Nguyen A, Ramesh A, Kumar S, Nandi D, Brouillard A, Wells A, Pobezinsky L, Osborne B, Kulkarni AA. Granzyme B nanoreporter for early monitoring of tumor response to immunotherapy. SCIENCE ADVANCES 2020; 6:6/40/eabc2777. [PMID: 33008894 PMCID: PMC7852386 DOI: 10.1126/sciadv.abc2777] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 08/14/2020] [Indexed: 05/08/2023]
Abstract
Despite recent advancements in cancer immunotherapy, accurate monitoring of its efficacy is challenging due to heterogeneous immune responses. Conventional imaging techniques lack the sensitivity and specificity for early response assessment. In this study, we designed a granzyme B (GrB) nanoreporter (GNR) that can deliver an immune checkpoint inhibitor to the tumor and track time-sensitive GrB activity as a direct way to monitor initiation of effective immune responses. Anti-programmed death-ligand 1 (PD-L1) antibody-conjugated GNRs inhibited PD-1/PD-L1 interactions efficiently and induced T cell-mediated GrB release that can be imaged using activatable imaging probe. GNRs enabled real-time immunotherapy response monitoring in a tumor-bearing mice model and distinguished between highly responsive and poorly responsive tumors. Furthermore, increasing doses resulted in a better response and enhanced sensitivity in poorly responsive tumors. These findings indicate that GNR has the potential to serve as a tool for sensitive and noninvasive evaluation of immunotherapy efficacy.
Collapse
Affiliation(s)
- Anh Nguyen
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, USA
| | - Anujan Ramesh
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA, USA
| | - Sahana Kumar
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, USA
| | - Dipika Nandi
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
| | - Anthony Brouillard
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, USA
| | - Alexandria Wells
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
| | - Leonid Pobezinsky
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
- Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, USA
| | - Barbara Osborne
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
- Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, USA
| | - Ashish A Kulkarni
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, USA.
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA, USA
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
- Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, USA
| |
Collapse
|
246
|
Thompson EA, Powell JD. Inhibition of the Adenosine Pathway to Potentiate Cancer Immunotherapy: Potential for Combinatorial Approaches. Annu Rev Med 2020; 72:331-348. [PMID: 32903139 DOI: 10.1146/annurev-med-060619-023155] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cancer immunotherapy has revolutionized the way that we think about treating cancer. Although checkpoint blockade therapy, including anti-PD-1/PD-L1 and anti-CTLA-4, has shown remarkable success, the responses are limited to only a subset of patients. This discrepancy highlights the many overlapping avenues for immune evasion or suppression that can be employed by a tumor. One such mechanism of immunosuppression is adenosinergic signaling within the tumor microenvironment. We provide an overview of the current status of clinical trials targeting the adenosine pathway, including CD73, CD39, and adenosine receptors. Additionally, we highlight several avenues that may be explored to further potentiate responses in the clinic by combining adenosine-targeting agents to target multiple arms of the pathway or by using conventional immunotherapy agents.
Collapse
Affiliation(s)
- Elizabeth A Thompson
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, USA; ,
| | - Jonathan D Powell
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, USA; ,
| |
Collapse
|
247
|
Centanni M, Moes DJAR, Trocóniz IF, Ciccolini J, van Hasselt JGC. Clinical Pharmacokinetics and Pharmacodynamics of Immune Checkpoint Inhibitors. Clin Pharmacokinet 2020; 58:835-857. [PMID: 30815848 PMCID: PMC6584248 DOI: 10.1007/s40262-019-00748-2] [Citation(s) in RCA: 222] [Impact Index Per Article: 44.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have demonstrated significant clinical impact in improving overall survival of several malignancies associated with poor outcomes; however, only 20–40% of patients will show long-lasting survival. Further clarification of factors related to treatment response can support improvements in clinical outcome and guide the development of novel immune checkpoint therapies. In this article, we have provided an overview of the pharmacokinetic (PK) aspects related to current ICIs, which include target-mediated drug disposition and time-varying drug clearance. In response to the variation in treatment exposure of ICIs and the significant healthcare costs associated with these agents, arguments for both dose individualization and generalization are provided. We address important issues related to the efficacy and safety, the pharmacodynamics (PD), of ICIs, including exposure–response relationships related to clinical outcome. The unique PK and PD aspects of ICIs give rise to issues of confounding and suboptimal surrogate endpoints that complicate interpretation of exposure–response analysis. Biomarkers to identify patients benefiting from treatment with ICIs have been brought forward. However, validated biomarkers to monitor treatment response are currently lacking.
Collapse
Affiliation(s)
- Maddalena Centanni
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Dirk Jan A R Moes
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Iñaki F Trocóniz
- Pharmacometrics and Systems Pharmacology, Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
| | - Joseph Ciccolini
- SMARTc, CRCM Inserm U1068 Aix Marseille Univ and La Timone University Hospital of Marseille, Marseille, France
| | - J G Coen van Hasselt
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands.
| |
Collapse
|
248
|
Eso Y, Seno H. Current status of treatment with immune checkpoint inhibitors for gastrointestinal, hepatobiliary, and pancreatic cancers. Therap Adv Gastroenterol 2020; 13:1756284820948773. [PMID: 32913444 PMCID: PMC7443993 DOI: 10.1177/1756284820948773] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 07/16/2020] [Indexed: 02/04/2023] Open
Abstract
The development of immune checkpoint inhibitors (ICIs) targeting cytotoxic T lymphocyte antigen 4 (CTLA-4), programmed cell death protein 1 (PD-1), or programmed cell death protein ligand 1 (PD-L1) has revolutionized the treatment strategy in various types of cancers. In addition, recent studies have revealed that tumor microsatellite instability (MSI) status and tumor mutation burden (TMB) contribute significantly to the therapeutic response to anti-PD-1 monoclonal antibody (mAb), which led to an accelerated approval to pembrolizumab for the treatment of MSI-high or mismatch-repair-deficient solid tumors after conventional chemotherapies in 2017 and for the treatment of TMB-high solid tumors in 2020 by the United States Food and Drug Administration (FDA). In the field of gastrointestinal cancers, many clinical trials evaluating the safety and efficacy of various regimens such as ICI monotherapy, the combination of anti-CTLA-4 mAb and anti-PD-1/PD-L1 mAb, and combination of ICI and conventional chemotherapy or tyrosine kinase inhibitor have been reported or are in progress. This review summarizes MSI status and TMB in gastrointestinal, hepatobiliary, and pancreatic cancers, and provides the results of most relevant clinical trials evaluating ICIs. We also discuss the development of biomarkers required for improving the selection of patients with a high probability of benefiting from treatment with ICIs, and potential therapeutic strategies that could help to enhance anticancer responses of ICIs.
Collapse
Affiliation(s)
| | - Hiroshi Seno
- Department of Gastroenterology and Hepatology,
Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
249
|
Wei WZ, Gibson HM, Jacob JB, Frelinger JA, Berzofsky JA, Maeng H, Dyson G, Reyes JD, Pilon-Thomas S, Ratner S, Wei KC. Diversity Outbred Mice Reveal the Quantitative Trait Locus and Regulatory Cells of HER2 Immunity. THE JOURNAL OF IMMUNOLOGY 2020; 205:1554-1563. [PMID: 32796024 DOI: 10.4049/jimmunol.2000466] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 07/11/2020] [Indexed: 01/14/2023]
Abstract
The genetic basis and mechanisms of disparate antitumor immune response was investigated in Diversity Outbred (DO) F1 mice that express human HER2. DO mouse stock samples nearly the entire genetic repertoire of the species. We crossed DO mice with syngeneic HER2 transgenic mice to study the genetics of an anti-self HER2 response in a healthy outbred population. Anti-HER2 IgG was induced by Ad/E2TM or naked pE2TM, both encoding HER2 extracellular and transmembrane domains. The response of DO F1 HER2 transgenic mice was remarkably variable. Still, immune sera inhibited HER2+ SKBR3 cell survival in a dose-dependent fashion. Using DO quantitative trait locus (QTL) analysis, we mapped the QTL that influences both total IgG and IgG2(a/b/c) Ab response to either Ad/E2TM or pE2TM. QTL from these four datasets identified a region in chromosome 17 that was responsible for regulating the response. A/J and NOD segments of genes in this region drove elevated HER2 Ig levels. This region is rich in MHC-IB genes, several of which interact with inhibitory receptors of NK cells. (B6xA/J)F1 and (B6xNOD)F1 HER2 transgenic mice received Ad/E2TM after NK cell depletion, and they produced less HER2 IgG, demonstrating positive regulatory function of NK cells. Depletion of regulatory T cells enhanced response. Using DO QTL analysis, we show that MHC-IB reactive NK cells exert positive influence on the immunity, countering negative regulation by regulatory T cells. This new, to our knowledge, DO F1 platform is a powerful tool for revealing novel immune regulatory mechanisms and for testing new interventional strategies.
Collapse
Affiliation(s)
- Wei-Zen Wei
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201;
| | - Heather M Gibson
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201
| | - Jennifer B Jacob
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201
| | - Jeffrey A Frelinger
- Valley Fever Center of Excellence, Department of Immunobiology, University of Arizona, Tucson, AZ 85724
| | - Jay A Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892; and
| | - Hoyoung Maeng
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892; and
| | - Gregory Dyson
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201
| | - Joyce D Reyes
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201
| | - Shari Pilon-Thomas
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612
| | - Stuart Ratner
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201
| | - Kuang-Chung Wei
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201
| |
Collapse
|
250
|
Vivarelli S, Falzone L, Grillo CM, Scandurra G, Torino F, Libra M. Cancer Management during COVID-19 Pandemic: Is Immune Checkpoint Inhibitors-Based Immunotherapy Harmful or Beneficial? Cancers (Basel) 2020; 12:E2237. [PMID: 32785162 PMCID: PMC7465907 DOI: 10.3390/cancers12082237] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 07/28/2020] [Accepted: 08/07/2020] [Indexed: 01/08/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) is currently representing a global health threat especially for fragile individuals, such as cancer patients. It was demonstrated that cancer patients have an increased risk of developing a worse symptomatology upon severe acute respiratory syndrome associated coronavirus-2 (SARS-CoV-2) infection, often leading to hospitalization and intensive care. The consequences of this pandemic for oncology are really heavy, as the entire healthcare system got reorganized. Both oncologists and cancer patients are experiencing rescheduling of treatments and disruptions of appointments with a concurrent surge of fear and stress. In this review all the up-to-date findings, concerning the association between COVID-19 and cancer, are reported. A remaining very debated question regards the use of an innovative class of anti-cancer molecules, the immune checkpoint inhibitors (ICIs), given their modulating effects on the immune system. For that reason, administration of ICIs to cancer patients represents a question mark during this pandemic, as its correlation with COVID-19-associated risks is still under investigation. Based on the mechanisms of action of ICIs and the current evidence, we suggest that ICIs not only can be safely administered to cancer patients, but they might even be beneficial in COVID-19-positive cancer patients, by exerting an immune-stimulating action.
Collapse
Affiliation(s)
- Silvia Vivarelli
- Laboratory of Translational Oncology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy;
| | - Luca Falzone
- Epidemiology Unit, IRCCS Istituto Nazionale Tumori ‘Fondazione G. Pascale’, I-80131 Naples, Italy
| | - Caterina Maria Grillo
- Otolaryngology Unit, Department of Medical Sciences, Surgical and Advanced Technologies, University of Catania, 95123 Catania, Italy;
| | - Giuseppa Scandurra
- Medical Oncology Unit, Azienda Ospedaliera Cannizzaro, 95126 Catania, Italy;
| | - Francesco Torino
- Department of Systems Medicine, Medical Oncology, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Massimo Libra
- Laboratory of Translational Oncology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy;
- Research Center for Prevention, Diagnosis and Treatment of Cancer, University of Catania, 95123 Catania, Italy
| |
Collapse
|