201
|
Buchbinder EI, Pfaff KL, Turner MM, Manos M, Ouyang O, Ott PA, Giobbie-Hurder A, Rodig SJ, Hodi FS. Brief Communication on Pathologic Assessment of Persistent Stable Metastatic Lesions in Patients Treated With Anti-CTLA-4 or Anti-CTLA-4 + Anti-PD-1 Therapy. J Immunother 2023; 46:192-196. [PMID: 37115942 PMCID: PMC10168111 DOI: 10.1097/cji.0000000000000470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 03/02/2023] [Indexed: 04/30/2023]
Abstract
Despite the wide use of immune checkpoint inhibition for the treatment of melanoma, the mechanisms leading to long-term stable disease are incompletely understood. Patients with metastatic melanoma who had received ipilimumab alone or ipilimumab plus nivolumab 2+years prior and attained at least 6 months of stable disease were identified. Positron emission tomography/computed tomography (PET/CT) was performed. Pretreatment and posttreatment biopsies of areas of stable disease were assessed for tumor, fibrosis, and inflammation. Seven patients underwent PET/CT and tissue biopsy. Fluorodeoxyglucose avid lesions on PET/CT ranged from no activity to an SUV of 22. In 6 patients, the residual stable lesions were composed of necrosis and fibrosis with a prominent pigment containing macrophages and no residual melanoma. In 1 patient, a nodal lesion demonstrated melanoma with active inflammation. In most patients with durable stable disease after treatment with ipilimumab or ipilimumab/nivolumab, residual lesions demonstrated predominantly necrosis and fibrosis consistent with resolving lesions. The presence of melanophages in these samples may suggest ongoing immune surveillance. One patient did demonstrate residual melanoma, indicating the need for ongoing monitoring of this patient population.
Collapse
Affiliation(s)
- Elizabeth I. Buchbinder
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Kathleen L. Pfaff
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Madison M. Turner
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Michael Manos
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Olivia Ouyang
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Patrick A. Ott
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA
- Harvard Medical School, Boston, MA
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Anita Giobbie-Hurder
- Division of Biostatistics, Department of Data Sciences, Dana-Farber Cancer Institute, Boston, MA
| | - Scott J. Rodig
- Harvard Medical School, Boston, MA
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - F. Stephen Hodi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA
- Harvard Medical School, Boston, MA
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA
| |
Collapse
|
202
|
Kelly CM, Qin LX, Whiting KA, Richards AL, Avutu V, Chan JE, Chi P, Dickson MA, Gounder MM, Keohan ML, Movva S, Nacev BA, Rosenbaum E, Adamson T, Singer S, Bartlett EK, Crago AM, Yoon SS, Hwang S, Erinjeri JP, Antonescu CR, Tap WD, D’Angelo SP. A Phase II Study of Epacadostat and Pembrolizumab in Patients with Advanced Sarcoma. Clin Cancer Res 2023; 29:2043-2051. [PMID: 36971773 PMCID: PMC10752758 DOI: 10.1158/1078-0432.ccr-22-3911] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/15/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023]
Abstract
PURPOSE Epacadostat, an indole 2,3 dioxygenase 1 (IDO1) inhibitor, proposed to shift the tumor microenvironment toward an immune-stimulated state, showed early promise in melanoma but has not been studied in sarcoma. This study combined epacadostat with pembrolizumab, which has modest activity in select sarcoma subtypes. PATIENTS AND METHODS This phase II study enrolled patients with advanced sarcoma into five cohorts including (i) undifferentiated pleomorphic sarcoma (UPS)/myxofibrosarcoma, (ii) liposarcoma (LPS), (iii) leiomyosarcoma (LMS), (iv) vascular sarcoma, including angiosarcoma and epithelioid hemangioendothelioma (EHE), and (v) other subtypes. Patients received epacadostat 100 mg twice daily plus pembrolizumab at 200 mg/dose every 3 weeks. The primary endpoint was best objective response rate (ORR), defined as complete response (CR) and partial response (PR), at 24 weeks by RECIST v.1.1. RESULTS Thirty patients were enrolled [60% male; median age 54 years (range, 24-78)]. The best ORR at 24 weeks was 3.3% [PR, n = 1 (leiomyosarcoma); two-sided 95% CI, 0.1%-17.2%]. The median PFS was 7.6 weeks (two-sided 95% CI, 6.9-26.7). Treatment was well tolerated. Grade 3 treatment-related adverse events occurred in 23% (n = 7) of patients. In paired pre- and post-treatment tumor samples, no association was found between treatment and PD-L1 or IDO1 tumor expression or IDO-pathway-related gene expression by RNA sequencing. No significant changes in serum tryptophan or kynurenine levels were observed after baseline. CONCLUSIONS Combination epacadostat and pembrolizumab was well tolerated and showed limited antitumor activity in sarcoma. Correlative analyses suggested that inadequate IDO1 inhibition was achieved.
Collapse
Affiliation(s)
- Ciara M. Kelly
- Department of Medicine, Memorial Sloan Kettering Cancer Center
- Department of Medicine, Weill Cornell Medical College
| | - Li-Xuan Qin
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center
| | - Karissa A. Whiting
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center
| | - Allison L. Richards
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center
| | - Viswatej Avutu
- Department of Medicine, Memorial Sloan Kettering Cancer Center
- Department of Medicine, Weill Cornell Medical College
| | - Jason E. Chan
- Department of Medicine, Memorial Sloan Kettering Cancer Center
- Department of Medicine, Weill Cornell Medical College
| | - Ping Chi
- Department of Medicine, Memorial Sloan Kettering Cancer Center
- Department of Medicine, Weill Cornell Medical College
| | - Mark A. Dickson
- Department of Medicine, Memorial Sloan Kettering Cancer Center
- Department of Medicine, Weill Cornell Medical College
| | - Mrinal M. Gounder
- Department of Medicine, Memorial Sloan Kettering Cancer Center
- Department of Medicine, Weill Cornell Medical College
| | - Mary Louise Keohan
- Department of Medicine, Memorial Sloan Kettering Cancer Center
- Department of Medicine, Weill Cornell Medical College
| | - Sujana Movva
- Department of Medicine, Memorial Sloan Kettering Cancer Center
- Department of Medicine, Weill Cornell Medical College
| | - Benjamin A. Nacev
- Department of Medicine, Memorial Sloan Kettering Cancer Center
- Department of Medicine, Weill Cornell Medical College
| | - Evan Rosenbaum
- Department of Medicine, Memorial Sloan Kettering Cancer Center
- Department of Medicine, Weill Cornell Medical College
| | - Travis Adamson
- Department of Medicine, Memorial Sloan Kettering Cancer Center
| | - Sam Singer
- Department of Surgery, Memorial Sloan Kettering Cancer Center
| | | | - Aimee M. Crago
- Department of Surgery, Memorial Sloan Kettering Cancer Center
| | - Sam S. Yoon
- Department of Surgery, Memorial Sloan Kettering Cancer Center
| | - Sinchun Hwang
- Department of Radiology, Memorial Sloan Kettering Cancer Center
| | | | | | - William D. Tap
- Department of Medicine, Memorial Sloan Kettering Cancer Center
- Department of Medicine, Weill Cornell Medical College
| | - Sandra P. D’Angelo
- Department of Medicine, Memorial Sloan Kettering Cancer Center
- Department of Medicine, Weill Cornell Medical College
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center
| |
Collapse
|
203
|
Huang C, Ren S, Chen Y, Liu A, Wu Q, Jiang T, Lv P, Song D, Hu F, Lan J, Sun L, Zheng X, Luo X, Chu Q, Jia K, Li Y, Wang J, Zou C, Hu J, Wang G. PD-L1 methylation restricts PD-L1/PD-1 interactions to control cancer immune surveillance. SCIENCE ADVANCES 2023; 9:eade4186. [PMID: 37235656 DOI: 10.1126/sciadv.ade4186] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 04/21/2023] [Indexed: 05/28/2023]
Abstract
Immune checkpoint inhibitors targeting programmed cell death protein 1 (PD-1) or programmed cell death 1 ligand 1 (PD-L1) have enabled some patients with cancer to experience durable, complete treatment responses; however, reliable anti-PD-(L)1 treatment response biomarkers are lacking. Our research found that PD-L1 K162 was methylated by SETD7 and demethylated by LSD2. Furthermore, PD-L1 K162 methylation controlled the PD-1/PD-L1 interaction and obviously enhanced the suppression of T cell activity controlling cancer immune surveillance. We demonstrated that PD-L1 hypermethylation was the key mechanism for anti-PD-L1 therapy resistance, investigated that PD-L1 K162 methylation was a negative predictive marker for anti-PD-1 treatment in patients with non-small cell lung cancer, and showed that the PD-L1 K162 methylation:PD-L1 ratio was a more accurate biomarker for predicting anti-PD-(L)1 therapy sensitivity. These findings provide insights into the regulation of the PD-1/PD-L1 pathway, identify a modification of this critical immune checkpoint, and highlight a predictive biomarker of the response to PD-1/PD-L1 blockade therapy.
Collapse
Affiliation(s)
- Changsheng Huang
- GI Cancer Research Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shengxiang Ren
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Yaqi Chen
- GI Cancer Research Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Anyi Liu
- GI Cancer Research Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qi Wu
- GI Cancer Research Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tao Jiang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Panjing Lv
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Da Song
- GI Cancer Research Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Fuqing Hu
- GI Cancer Research Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jingqing Lan
- GI Cancer Research Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Li Sun
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xue Zheng
- Wuhan Blood Center, Wuhan 430030, China
| | - Xuelai Luo
- GI Cancer Research Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qian Chu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Keyi Jia
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Yan Li
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jun Wang
- Department of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Caicun Zou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Junbo Hu
- GI Cancer Research Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Guihua Wang
- GI Cancer Research Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
204
|
Karnik I, Her Z, Neo SH, Liu WN, Chen Q. Emerging Preclinical Applications of Humanized Mouse Models in the Discovery and Validation of Novel Immunotherapeutics and Their Mechanisms of Action for Improved Cancer Treatment. Pharmaceutics 2023; 15:1600. [PMID: 37376049 DOI: 10.3390/pharmaceutics15061600] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 05/22/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Cancer therapeutics have undergone immense research over the past decade. While chemotherapies remain the mainstay treatments for many cancers, the advent of new molecular techniques has opened doors for more targeted modalities towards cancer cells. Although immune checkpoint inhibitors (ICIs) have demonstrated therapeutic efficacy in treating cancer, adverse side effects related to excessive inflammation are often reported. There is a lack of clinically relevant animal models to probe the human immune response towards ICI-based interventions. Humanized mouse models have emerged as valuable tools for pre-clinical research to evaluate the efficacy and safety of immunotherapy. This review focuses on the establishment of humanized mouse models, highlighting the challenges and recent advances in these models for targeted drug discovery and the validation of therapeutic strategies in cancer treatment. Furthermore, the potential of these models in the process of uncovering novel disease mechanisms is discussed.
Collapse
Affiliation(s)
- Isha Karnik
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
| | - Zhisheng Her
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Shu Hui Neo
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Wai Nam Liu
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Qingfeng Chen
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| |
Collapse
|
205
|
Song SH, Ghosh T, You DG, Joo H, Lee J, Lee J, Kim CH, Jeon J, Shin S, Park JH. Functionally Masked Antibody to Uncouple Immune-Related Toxicities in Checkpoint Blockade Cancer Therapy. ACS NANO 2023. [PMID: 37184643 DOI: 10.1021/acsnano.2c12532] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Of the existing immunotherapy drugs in oncology, monoclonal antibodies targeting the immune checkpoint axis are preferred because of the durable responses observed in selected patients. However, the associated immune-related adverse events (irAEs), causing uncommon fatal events, often require specialized management and medication discontinuation. The study aim was to investigate our hypothesis that masking checkpoint antibodies with tumor microenvironment (TME)-responsive polymer chains can mitigate irAEs and selectively target tumors by limiting systemic exposure to patients. We devised a broadly applicable strategy that functionalizes immune checkpoint-blocking antibodies with a mildly acidic pH-cleavable poly(ethylene glycol) (PEG) shell to prevent inflammatory side effects in normal tissues. Conjugation of pH-sensitive PEG to anti-CD47 antibodies (αCD47) minimized antibody-cell interactions by inhibiting their binding ability and functionality at physiological pH, leading to prevention of αCD47-induced anemia in tumor-bearing mice. When conjugated to anti-CTLA-4 and anti-PD-1 antibodies, double checkpoint blockade-induced colitis was also ameliorated. Notably, removal of the protective shell in response to an acidic TME restored the checkpoint antibody activities, accompanied by effective tumor regression and long-term survival in the mouse model. Our results support a feasible strategy for antibody-based therapies to uncouple toxicity from efficacy and show the translational potential for cancer immunotherapy.
Collapse
Affiliation(s)
- Seok Ho Song
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Torsha Ghosh
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Dong Gil You
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Hyeyeon Joo
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jeongjin Lee
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Republic of Korea
| | - Jaeah Lee
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Chan Ho Kim
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jueun Jeon
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Sol Shin
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Republic of Korea
| | - Jae Hyung Park
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Republic of Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
206
|
Gilbert D, Hu J, Medina T, Kessler ER, Lam ET. Safety of COVID-19 vaccines in subjects with solid tumor cancers receiving immune checkpoint inhibitors. Hum Vaccin Immunother 2023:2207438. [PMID: 37157982 PMCID: PMC10294768 DOI: 10.1080/21645515.2023.2207438] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023] Open
Abstract
The incidence of severe immune-related adverse events (irAEs) in cancer subjects receiving immune checkpoint inhibitors (ICIs) following COVID-19 vaccination and the relationship between the incidence of severe irAE and the interval between COVID-19 vaccination and ICI dose have not been established. We performed a retrospective study evaluating the incidence of irAEs in solid tumor subjects receiving ICI therapy who received any COVID-19 vaccinations since FDA authorization. irAEs were defined as severe with one or more grade 3 or above events (CTCAE v5.0), multiple organ involvement, or requiring hospitalization for management. Two hundred and eighty-four subjects who received COVID vaccinations from December 2020 and February 2022 were included in this analysis [median age at vaccination 67 years (IQR 59.0-75.0); 67.3% male]. Twenty-nine subjects (10.2%) developed severe irAEs, of which 12 subjects (41.4%) received ICI monotherapy, 10 subjects (34.5%) received combination ICI therapy with nivolumab and ipilimumab, and 7 subjects (24.1%) received ICI plus VEGFR-TKI therapy. Hospitalization occurred in 62% of subjects with severe irAEs, with a median duration of 3 days (IQR: 3.0-7.5 days). Immunosuppressive therapy was required in 79.3%, with a median duration of 103 days (IQR: 42.0-179.0). ICI therapy was discontinued in 51.7% of subjects with severe irAE; dosing was held or interrupted in 34.5%. Among severe irAEs, the median interval between vaccination and ICI treatment closest to the occurrence of severe irAE was 15.5 days (IQR: 10.0-23.0). In solid tumor cancer subjects receiving ICIs, COVID-19 vaccination is not associated with an increased incidence of severe irAEs compared to historical data and may be safely administered during ICI cancer therapy in subjects who lack contraindications.
Collapse
Affiliation(s)
- Danielle Gilbert
- Department of Internal Medicine, Division of Medical Oncology, University of Colorado Cancer Center, University of Anschutz Medical Campus, Aurora, CO, USA
| | - Junxiao Hu
- Department of Biostatistics, University of Colorado Cancer Center Biostatistics Core, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Theresa Medina
- Department of Internal Medicine, Division of Medical Oncology, University of Colorado Cancer Center, University of Anschutz Medical Campus, Aurora, CO, USA
| | - Elizabeth R Kessler
- Department of Internal Medicine, Division of Medical Oncology, University of Colorado Cancer Center, University of Anschutz Medical Campus, Aurora, CO, USA
| | - Elaine T Lam
- Department of Internal Medicine, Division of Medical Oncology, University of Colorado Cancer Center, University of Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
207
|
Zhao Y, Ma Y, Zang A, Cheng Y, Zhang Y, Wang X, Chen Z, Qu S, He J, Chen C, Jin C, Zhu D, Li Q, Liu X, Su W, Ba Y, Hao Y, Chen J, Zhang G, Qu S, Li Y, Feng W, Yang M, Liu B, Ouyang W, Liang J, Yu Z, Kang X, Xue S, Yang G, Yan W, Yang Y, Liu Z, Peng Y, Fanslow B, Huang X, Zhang L, Zhao H. First-in-human phase I/Ib study of QL1706 (PSB205), a bifunctional PD1/CTLA4 dual blocker, in patients with advanced solid tumors. J Hematol Oncol 2023; 16:50. [PMID: 37158938 PMCID: PMC10169367 DOI: 10.1186/s13045-023-01445-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 04/26/2023] [Indexed: 05/10/2023] Open
Abstract
BACKGROUND QL1706 (PSB205) is a single bifunctional MabPair (a novel technical platform) product consisting of two engineered monoclonal antibodies (anti-PD-1 IgG4 and anti-CTLA-4 IgG1), with a shorter elimination half-life (t1/2) for CTLA-4. We report results from a phase I/Ib study of QL1706 in patients with advanced solid tumors who failed standard therapies. METHODS In the phase I study, QL1706 was administered intravenously once every 3 weeks at one of five doses ranging from 0.3 to 10 mg/kg, and the maximum tolerated dose, recommended phase 2 dose (RP2D), safety, pharmacokinetics (PK), and pharmacodynamics (PD) of QL1706 were investigated. In the phase Ib study, QL1706 was administered at the RP2D intravenously every 3 weeks, and the preliminary efficacies in non-small cell lung cancer (NSCLC), nasopharyngeal carcinoma (NPC), cervical cancer (CC), and other solid tumors were evaluated. RESULTS Between March 2020 and July 2021, 518 patients with advanced solid tumors were enrolled (phase I, n = 99; phase Ib, n = 419). For all patients, the three most common treatment-related adverse events (TRAEs) were rash (19.7%), hypothyroidism (13.5%), and pruritus (13.3%). The TRAEs and immune-related adverse events (irAEs) of grade ≥ 3 occurred in 16.0% and 8.1% of patients, respectively. In phase I, 2 of 6 patients in the 10mg/kg group experienced dose-limiting toxicities (DLTs) (grade 3 thrombocytopenia and grade 4 immune-mediated nephritis), so the maximum tolerated dose (MTD) was reached at 10 mg/kg. The RP2D was determined to be 5 mg/kg based on comprehensive analysis of tolerability, PK/PD, and efficacy. For all patients who received QL1706 at the RP2D, the objective response rate (ORR) and median duration of response were 16.9% (79/468) and 11.7 months (8.3-not reached [NR]), respectively; and the ORRs were 14.0% (17/121) in NSCLC, 24.5% (27/110) in NPC, 27.3% (15/55) in CC, 7.4% (2/27) in colorectal cancer, 23.1% (6/26) in small cell lung cancer. For immunotherapy-naive patients, QL1706 exhibited promising antitumor activities, especially in NSCLC, NPC, and CC, with ORRs of 24.2%, 38.7%, and 28.3%, respectively. CONCLUSIONS QL1706 was well tolerated and demonstrated promising antitumor activity in solid tumors, especially in NSCLC, NPC, and CC patients. It is currently being evaluated in randomized phase II (NCT05576272, NCT05179317) and phase III (NCT05446883, NCT05487391) trials. Trial Registration ClinicalTrials.gov Identifier: NCT04296994 and NCT05171790.
Collapse
Affiliation(s)
- Yuanyuan Zhao
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, No. 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Yuxiang Ma
- Department of Clinical Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, No. 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Aimin Zang
- Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding, 071000, China
| | - Ying Cheng
- Department of Thoracic Oncology, Jilin Cancer Hospital, Changchun, 130012, China
| | - Yiping Zhang
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Xiangcai Wang
- Department of Oncology, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341001, China
| | - Zhendong Chen
- Department of Medical Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230093, China
| | - Song Qu
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Cancer Institute of Guangxi, Nanning, 530021, Guangxi, China
| | - Jianbo He
- Department of Medical Oncology of Respiratory, Guangxi Medical University Cancer Hospital, Cancer Institute of Guangxi, Nanning, 530021, Guangxi, China
| | - Chuanben Chen
- Department of Head and Neck Radiation Oncology, Fujian Cancer Hospital, Fuzhou, 350000, China
| | - Chuan Jin
- Department of Medical Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, 510095, China
| | - Dongyuan Zhu
- Rare Tumors Department, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Qingshan Li
- Department of Oncology, Affiliated Hospital of Chengde Medical University, Chengde, 067000, China
| | - Xianling Liu
- Department of Oncology, Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Wuyun Su
- Department of Medical Oncology, Affiliated Hospital of Inner Mongolia Medical University, Huhhot, 010050, Inner Mongolia, China
| | - Yi Ba
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Yanrong Hao
- Department of Oncology, Clinical Oncology Center, The People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, 530021, China
| | - Junmin Chen
- Department of Medical Oncology, Hainan General Hospital, Haikou, 570100, China
| | - Guoping Zhang
- Department of Medical Oncology, Yuebei People's Hospital, Shaoguan, 512025, China
| | - Shenhong Qu
- Department of Otolaryngology & Head and Neck, The People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, 530021, China
| | - Yong Li
- Department of Medical Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, 330000, China
| | - Weineng Feng
- Department of Head and Neck/Thoracic Medical Oncology, The First People's Hospital of Foshan, Foshan City, 528010, China
| | - Mengxiang Yang
- Oncology Department, Liaocheng People's Hospital, Liaocheng, 252004, China
| | - Baorui Liu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University and Clinical Cancer Institute of Nanjing University, Nanjing, 210008, China
| | - Weiwei Ouyang
- Department of Oncology, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, 550001, China
| | - Jin Liang
- Department of Oncology, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Zhuang Yu
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Xiaoyan Kang
- Clinical Research Center, Qilu Pharmaceutical Co., Ltd., Jinan, 250000, China
| | - Shilin Xue
- Clinical Research Center, Qilu Pharmaceutical Co., Ltd., Jinan, 250000, China
| | - Guihong Yang
- Department of Clinical Pharmacology, Qilu Pharmaceutical Co., Ltd., Jinan, 250000, China
| | - Wei Yan
- Sound Biologics, 21720 23rd Drive SE, Suite200, Bothell, WA, 98021, USA
| | - Yingying Yang
- Department of Non-Clinical, Qilu Pharmaceutical Co., Ltd., Jinan, 250001, China
| | - Zhi Liu
- Sound Biologics, 21720 23rd Drive SE, Suite200, Bothell, WA, 98021, USA
| | - Yufeng Peng
- Sound Biologics, 21720 23rd Drive SE, Suite200, Bothell, WA, 98021, USA
| | - Bill Fanslow
- Sound Biologics, 21720 23rd Drive SE, Suite200, Bothell, WA, 98021, USA
| | - Xian Huang
- Sound Biologics, 21720 23rd Drive SE, Suite200, Bothell, WA, 98021, USA
| | - Li Zhang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, No. 651 Dongfeng East Road, Guangzhou, 510060, China.
| | - Hongyun Zhao
- Department of Clinical Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, No. 651 Dongfeng East Road, Guangzhou, 510060, China.
| |
Collapse
|
208
|
Miah A, Tinoco G, Zhao S, Wei L, Johns A, Patel S, Li M, Grogan M, Lopez G, Husain M, Hoyd R, Mumtaz K, Meara A, Bertino EM, Kendra K, Spakowicz D, Otterson GA, Presley CJ, Owen DH. Immune checkpoint inhibitor-induced hepatitis injury: risk factors, outcomes, and impact on survival. J Cancer Res Clin Oncol 2023; 149:2235-2242. [PMID: 36070148 PMCID: PMC10191203 DOI: 10.1007/s00432-022-04340-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/29/2022] [Indexed: 10/14/2022]
Abstract
PURPOSE Immune checkpoint inhibitors (ICIs) are associated with a unique set of immune-related adverse events (irAEs). Few studies have evaluated the risk factors and outcomes of patients who develop ICI-induced hepatitis (ICIH). METHODS We utilized an institutional database of patients with advanced cancers treated with ICI to identify patients with ICIH. irAEs were graded using the Common Terminology Criteria for Adverse Events v4. Overall survival (OS) was calculated from the date of ICI to death from any cause or the date of the last follow-up. OS with 95% confidence intervals were estimated using the Kaplan-Meier method and stratified by the occurrence of ICIH. RESULTS We identified 1096 patients treated with ICI. The most common ICIs were PD1/L1 (n = 774) and CTLA-4 inhibitors (n = 195). ICIH occurred among 64 (6%) patients: severity was < grade 3 in 30 and ≥ grade 3 in 24 patients (3.1% overall). Median time to ICIH was 63 days. ICIH was more frequent in women (p = 0.038), in patients treated with combination ICIs (p < 0.001), and when given as first-line therapy (p = 0.018). Occurrence of ICIH was associated with significantly longer OS, median 37.0 months (95% CI 21.4, NR) compared to 11.3 months (95% CI 10, 13, p < 0.001); there was no difference in OS between patients with ≥ grade 3 ICIH vs grade 1-2. CONCLUSIONS Female sex, combination immunotherapy, and the first line of immunotherapy were associated with ICIH. Patients with ICIH had improved clinical survival compared to those that did not develop ICIH. There is a need for prospective further studies to confirm our findings.
Collapse
Affiliation(s)
- Abdul Miah
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, 1810 Cannon Drive, Suite 1240C, Columbus, OH, 43210, USA
| | - Gabriel Tinoco
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, 1810 Cannon Drive, Suite 1240C, Columbus, OH, 43210, USA.
| | - Songzhu Zhao
- Department of Biomedical Informatics and Center for Biostatistics, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Lai Wei
- Department of Biomedical Informatics and Center for Biostatistics, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Andrew Johns
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, 1810 Cannon Drive, Suite 1240C, Columbus, OH, 43210, USA
| | - Sandip Patel
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, 1810 Cannon Drive, Suite 1240C, Columbus, OH, 43210, USA
| | - Mingjia Li
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, 1810 Cannon Drive, Suite 1240C, Columbus, OH, 43210, USA
| | - Madison Grogan
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, 1810 Cannon Drive, Suite 1240C, Columbus, OH, 43210, USA
| | - Gabrielle Lopez
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, 1810 Cannon Drive, Suite 1240C, Columbus, OH, 43210, USA
| | - Marium Husain
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, 1810 Cannon Drive, Suite 1240C, Columbus, OH, 43210, USA
| | - Rebecca Hoyd
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, 1810 Cannon Drive, Suite 1240C, Columbus, OH, 43210, USA
| | - Khalid Mumtaz
- Division of Gastroenterology Hepatology and Nutrition, The Ohio State University, Columbus, OH, USA
| | - Alexa Meara
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, 1810 Cannon Drive, Suite 1240C, Columbus, OH, 43210, USA
- Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Erin M Bertino
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, 1810 Cannon Drive, Suite 1240C, Columbus, OH, 43210, USA
| | - Kari Kendra
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, 1810 Cannon Drive, Suite 1240C, Columbus, OH, 43210, USA
| | - Daniel Spakowicz
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, 1810 Cannon Drive, Suite 1240C, Columbus, OH, 43210, USA
| | - Gregory A Otterson
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, 1810 Cannon Drive, Suite 1240C, Columbus, OH, 43210, USA
| | - Carolyn J Presley
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, 1810 Cannon Drive, Suite 1240C, Columbus, OH, 43210, USA
| | - Dwight H Owen
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, 1810 Cannon Drive, Suite 1240C, Columbus, OH, 43210, USA
| |
Collapse
|
209
|
Sah VR, Jespersen H, Karlsson J, Nilsson LM, Bergqvist M, Johansson I, Carneiro A, Helgadottir H, Levin M, Ullenhag G, Ståhlberg A, Olofsson Bagge R, Nilsson JA, Ny L. Chemokine Analysis in Patients with Metastatic Uveal Melanoma Suggests a Role for CCL21 Signaling in Combined Epigenetic Therapy and Checkpoint Immunotherapy. CANCER RESEARCH COMMUNICATIONS 2023; 3:884-895. [PMID: 37377898 PMCID: PMC10194136 DOI: 10.1158/2767-9764.crc-22-0490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 03/01/2023] [Accepted: 04/28/2023] [Indexed: 06/29/2023]
Abstract
Purpose Patients with metastatic uveal melanoma have limited therapeutic options and high mortality rate so new treatment options are needed. Patients and Methods We previously reported that patients treated with the PD-1 inhibitor pembrolizumab and the histone deacetylase inhibitor entinostat in the PEMDAC trial, experienced clinical benefits if their tumor originated from iris or was wildtype for BAP1 tumor suppressor gene. Here we present the 2-year follow-up of the patients in the PEMDAC trial and identify additional factors that correlate with response or survival. Results Durable responses were observed in 4 patients, with additional 8 patients exhibiting a stable disease. The median overall survival was 13.7 months. Grade 3 adverse events were reported in 62% of the patients, but they were all manageable. No fatal toxicity was observed. Activity of thymidine kinase 1 in plasma was higher in patients with stable disease or who progressed on treatment, compared with those with partial response. Chemokines and cytokines were analyzed in plasma. Three chemokines were significantly different when comparing patients with and without response. One of the factors, CCL21, was higher in the plasma of responding patients before treatment initiation but decreased in the same patients upon treatment. In tumors, CCL21 was expressed in areas resembling tertiary lymphoid structures (TLS). High plasma levels of CCL21 and presence of TLS-like regions in the tumor correlated with longer survival. Conclusions This study provides insight into durable responses in the PEMDAC trial, and describes dynamic changes of chemokines and cytokines in the blood of these patients. Significance The most significant finding from the 2-year follow-up study of the PEMDAC trial was that high CCL21 levels in blood was associated with response and survival. CCL21 was also expressed in TLS-like regions and presence of these regions was associated with longer survival. These analyses of soluble and tumor markers can inform on predictive biomarkers needing validation and become hypothesis generating for experimental research.
Collapse
Affiliation(s)
- Vasu R. Sah
- Sahlgrenska Center for Cancer Research, Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Jespersen
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Joakim Karlsson
- Harry Perkins Institute of Medical Research, University of Western Australia, Perth, Western Australia, Australia
| | - Lisa M. Nilsson
- Harry Perkins Institute of Medical Research, University of Western Australia, Perth, Western Australia, Australia
| | | | - Iva Johansson
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ana Carneiro
- Department of Oncology, Skåne University Hospital, Lund, Sweden
| | - Hildur Helgadottir
- Department of Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Max Levin
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Gustav Ullenhag
- Department of Radiology, Oncology and Radiation Science, Section of Oncology, Uppsala University, Uppsala, Sweden
| | - Anders Ståhlberg
- Department of Laboratory Medicine, Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Genetics and Genomics, Sahlgrenska Center for Cancer Research, Institute of Biomedicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Roger Olofsson Bagge
- Sahlgrenska Center for Cancer Research, Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Jonas A. Nilsson
- Sahlgrenska Center for Cancer Research, Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Harry Perkins Institute of Medical Research, University of Western Australia, Perth, Western Australia, Australia
| | - Lars Ny
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
210
|
Ma P, Liu J, Qin J, Lai L, Heo GS, Luehmann H, Sultan D, Bredemeyer A, Bajapa G, Feng G, Jimenez J, Parks A, Amrute J, Villanueva A, Liu Y, Lin CY, Mack M, Amancherla K, Moslehi J, Lavine KJ. Expansion of Disease Specific Cardiac Macrophages in Immune Checkpoint Inhibitor Myocarditis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.28.538426. [PMID: 37162929 PMCID: PMC10168426 DOI: 10.1101/2023.04.28.538426] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Background Immune checkpoint inhibitors (ICIs), antibodies targeting PD-1/PD-L1 or CTLA4 have revolutionized cancer management but are associated with devastating immune-related adverse events (irAEs) including myocarditis. The main risk factor for ICI myocarditis is the use of combination PD-1 and CTLA4 inhibition. ICI-myocarditis is often fulminant and is pathologically characterized by myocardial infiltration of T lymphocytes and macrophages. While much has been learned regarding the role of T-cells in ICI-myocarditis, little is understood regarding the identity, transcriptional diversity, and functions of infiltrating macrophages. Methods We employed an established murine ICI myocarditis model ( Ctla4 +/- Pdcd1 -/- mice) to explore the cardiac immune landscape using single-cell RNA-sequencing, immunostaining, flow cytometry, in situ RNA hybridization and molecular imaging and antibody neutralization studies. Results We observed marked increases in CCR2 + monocyte-derived macrophages and CD8 + T-cells in this model. The macrophage compartment was heterogeneous and displayed marked enrichment in an inflammatory CCR2 + subpopulation highly expressing Cxcl9 , Cxcl10 , Gbp2b , and Fcgr4 that originated from CCR2 + monocytes. Importantly, a similar macrophage population expressing CXCL9 , CXCL10 , and CD16α (human homologue of mouse FcgR4) was found selectively expanded in patients with ICI myocarditis compared to other forms of heart failure and myocarditis. In silico prediction of cell-cell communication suggested interactions between T-cells and Cxcl9 + Cxcl10 + macrophages via IFN-γ and CXCR3 signaling pathways. Depleting CD8 + T-cells, macrophages, and blockade of IFN-γ signaling blunted the expansion of Cxcl9 + Cxcl10 + macrophages in the heart and attenuated myocarditis suggesting that this interaction was necessary for disease pathogenesis. Conclusion These data demonstrate that ICI-myocarditis is associated with the expansion of a specific population of IFN-γ induced inflammatory macrophages and suggest the possibility that IFN-γ blockade may be considered as a treatment option for this devastating condition.
Collapse
Affiliation(s)
- Pan Ma
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jing Liu
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Juan Qin
- Division of Cardiology, Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Lulu Lai
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Gyu Seong Heo
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Hannah Luehmann
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Deborah Sultan
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Andrea Bredemeyer
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Geetika Bajapa
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Guoshuai Feng
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jesus Jimenez
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Antanisha Parks
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Junedh Amrute
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Ana Villanueva
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Yongjian Liu
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Chieh-Yu Lin
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Matthias Mack
- Department of Internal Medicine II – Nephrology, Universitatsklinikum Regensburg Klinik und Poliklinik Innere Medizin II, Regensburg, Bayern, Germany
| | - Kaushik Amancherla
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Javid Moslehi
- Division of Cardiology, Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Kory J. Lavine
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
| |
Collapse
|
211
|
Musha A, Kubo N, Kawamura H, Okano N, Sato H, Okada K, Tomizawa K, Ota N, Adachi A, Shino M, Nikkuni O, Ida S, Shirai K, Saitoh JI, Yokoo S, Chikamatsu K, Ohno T. Efficacy of immune checkpoint inhibitor treatment for head and neck mucosal melanoma recurrence in patients treated with carbon-ion radiotherapy. Cancer Rep (Hoboken) 2023:e1825. [PMID: 37115713 PMCID: PMC10363791 DOI: 10.1002/cnr2.1825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/15/2023] [Accepted: 04/10/2023] [Indexed: 04/29/2023] Open
Abstract
BACKGROUND Carbon-ion radiotherapy (C-ion RT) is effective for head and neck mucosal melanoma (HN-MM), including radioresistant mucosal melanoma. Melanoma also responds effectively to immune checkpoint inhibitors (ICIs). Data on the efficacy and safety of ICIs for HN-MM are insufficient. AIMS To analyze the efficacy and safety of ICI salvage therapy in patients with HN-MM recurrence after C-ion RT. METHODS AND RESULTS This retrospective study analyzed the medical records of 52 patients with HN-MM treated with C-ion RT between 2012 and 2020. A dose of 57.6 or 64.0 Gy (relative biological effectiveness) was provided in 16 fractions. The primary endpoint was 3-year overall survival (OS) rate. The median follow-up time was 26.8 months for all patients. A total of 29 patients had local recurrence or distant metastasis, and 16 patients who received ICI therapy. The 3-year OS rate in the ICI group (n = 16) and best supportive care group (n = 13) were 53.8% and 0.0%, respectively (p = 0.837); the difference was not statistically significant. There were no deaths after 1 year among patients who underwent ICI therapy. No adverse events associated with C-ion RT were related to or exacerbated by ICI. CONCLUSION ICI salvage therapy is effective and safe for patients with HN-MM recurrence after C-ion RT.
Collapse
Affiliation(s)
- Atsushi Musha
- Gunma University Heavy Ion Medical Center, Maebashi, Japan
- Department of Oral and Maxillofacial Surgery and Plastic Surgery, Gunma University Graduate School of Medicine, Maebashi, Maebashi, Japan
| | - Nobuteru Kubo
- Gunma University Heavy Ion Medical Center, Maebashi, Japan
| | | | - Naoko Okano
- Gunma University Heavy Ion Medical Center, Maebashi, Japan
| | - Hiro Sato
- Gunma University Heavy Ion Medical Center, Maebashi, Japan
| | - Kohei Okada
- Gunma University Heavy Ion Medical Center, Maebashi, Japan
| | - Kento Tomizawa
- Gunma University Heavy Ion Medical Center, Maebashi, Japan
| | - Norichika Ota
- Gunma University Heavy Ion Medical Center, Maebashi, Japan
| | - Akiko Adachi
- Gunma University Heavy Ion Medical Center, Maebashi, Japan
| | - Masato Shino
- Department of Otolaryngology-Head and Neck Surgery, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Osamu Nikkuni
- Department of Otolaryngology-Head and Neck Surgery, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Shota Ida
- Department of Otolaryngology-Head and Neck Surgery, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Katsuyuki Shirai
- Gunma University Heavy Ion Medical Center, Maebashi, Japan
- Department of Radiology, Jichi Medical University Hospital, Tochigi, Japan
| | - Jun-Ichi Saitoh
- Gunma University Heavy Ion Medical Center, Maebashi, Japan
- Division of Radiation Oncology, Department of Radiology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Satoshi Yokoo
- Department of Oral and Maxillofacial Surgery and Plastic Surgery, Gunma University Graduate School of Medicine, Maebashi, Maebashi, Japan
| | - Kazuaki Chikamatsu
- Department of Otolaryngology-Head and Neck Surgery, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Tatsuya Ohno
- Gunma University Heavy Ion Medical Center, Maebashi, Japan
| |
Collapse
|
212
|
Rogers JE, Yamashita K, Sewastjanow-Silva M, Rosa Vicentini E, Waters R, Ajani JA. Nivolumab combination therapy as first-line treatments for unresectable, advanced or metastatic esophageal squamous cell carcinoma. Expert Rev Anticancer Ther 2023; 23:565-571. [PMID: 37122102 DOI: 10.1080/14737140.2023.2207826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
INTRODUCTION Esophageal cancers continue to confer a dismal prognosis. Targeted and immune therapies have skyrocketed in the world of cancer management. Unlike other solid tumors, esophageal squamous cell carcinoma (ESCC) has lacked effective targeted therapy. Promising outcomes with immune checkpoint inhibitors (ICIs) have recently changed ESCC management. AREAS COVERED Nivolumab has been granted several approvals to treat ESCC patients. Nivolumab is recommended as adjuvant therapy for localized ESCC patients following trimodality therapy who have residual cancer in the surgical specimen (lymph node(s) and or the primary). CheckMate-648 led to dual ICI therapy approval with nivolumab plus ipilimumab or nivolumab plus platinum with fluoropyrimidine as first line treatment for unresectable ESCC patients. ATTRACTION-3 resulted in nivolumab approval for second line therapy of unresectable ESCC patients who have not been exposed to ICI. Here we provide a review of nivolumab and how this relates to ESCC management. EXPERT OPINION Some ESCC patients will not experience a response to ICIs. Determining intrinsic and acquired resistance patterns are needed to further capitalize on ICI therapy for ESCC patients. PD-L1 expression has been explored as a potential biomarker. Data shows, however, PD-L1 positive tumor patients benefit but this assessment is not always needed.
Collapse
Affiliation(s)
- Jane E Rogers
- Departments of Pharmacy Clinical Programs, University of Texas MD Anderson Cancer Center, Texas, USA
| | - Kohei Yamashita
- Gastrointestinal Medical Oncology1, University of Texas MD Anderson Cancer Center, Texas, USA
| | | | - Ernesto Rosa Vicentini
- Gastrointestinal Medical Oncology1, University of Texas MD Anderson Cancer Center, Texas, USA
| | - Rebecca Waters
- Department of Pathology, University of Texas MD Anderson Cancer Center, Texas, USA
| | - Jaffer A Ajani
- Gastrointestinal Medical Oncology1, University of Texas MD Anderson Cancer Center, Texas, USA
| |
Collapse
|
213
|
Nahar S, Huang Y, Nagy BA, Zebala JA, Maeda DY, Rudloff U, Oppenheim JJ, Yang D. Regression and Eradication of Triple-Negative Breast Carcinoma in 4T1 Mouse Model by Combination Immunotherapies. Cancers (Basel) 2023; 15:cancers15082366. [PMID: 37190294 DOI: 10.3390/cancers15082366] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 04/07/2023] [Accepted: 04/17/2023] [Indexed: 05/17/2023] Open
Abstract
Triple-negative breast carcinoma (TNBC) is one of the most aggressive types of solid-organ cancers. While immune checkpoint blockade (ICB) therapy has significantly improved outcomes in certain types of solid-organ cancers, patients with immunologically cold TNBC are afforded only a modest gain in survival by the addition of ICB to systemic chemotherapy. Thus, it is urgently needed to develop novel effective therapeutic approaches for TNBC. Utilizing the 4T1 murine model of TNBC, we developed a novel combination immunotherapeutic regimen consisting of intratumoral delivery of high-mobility group nucleosome binding protein 1 (HMGN1), TLR2/6 ligand fibroblast-stimulating lipopeptide (FSL-1), TLR7/8 agonist (R848/resiquimod), and CTLA-4 blockade. We also investigated the effect of adding SX682, a small-molecule inhibitor of CXCR1/2 known to reduce MDSC trafficking to tumor microenvironment, to our therapeutic approach. 4T1-bearing mice responded with significant tumor regression and tumor elimination to our therapeutic combination regimen. Mice with complete tumor regressions did not recur and became long-term survivors. Treatment with HMGN1, FSL-1, R848, and anti-CTLA4 antibody increased the number of infiltrating CD4+ and CD8+ effector/memory T cells in both tumors and draining lymph nodes and triggered the generation of 4T1-specific cytotoxic T lymphocytes (CTLs) in the draining lymph nodes. Thus, we developed a potentially curative immunotherapeutic regimen consisting of HMGN1, FSL-1, R848, plus a checkpoint inhibitor for TNBC, which does not rely on the administration of chemotherapy, radiation, or exogenous tumor-associated antigen(s).
Collapse
Affiliation(s)
- Saifun Nahar
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, USA
- Rare Tumor Initiative, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Yue Huang
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| | - Bethany A Nagy
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| | | | | | - Udo Rudloff
- Rare Tumor Initiative, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Joost J Oppenheim
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| | - De Yang
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| |
Collapse
|
214
|
Zhang F, Guangchuan W, Chow R, He E, Majety M, Zhang Y, Chen S. Multiplexed inhibition of immunosuppressive genes with Cas13d for on-demand combinatorial cancer immunotherapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.14.532668. [PMID: 36993222 PMCID: PMC10055084 DOI: 10.1101/2023.03.14.532668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2023]
Abstract
Checkpoint blockade immunotherapy is a potent class of cancer treatment, however, the complex immunosuppressive tumor microenvironment (TME) often requires multi-agent combinations to be effective. Current cancer immunotherapy combination approaches are cumbersome, usually involving one-drug-at-a-time scheme. Here, we devise Multiplex Universal Combinatorial Immunotherapy via Gene-silencing (MUCIG), as a versatile approach for combinatorial cancer immunotherapy. We harness CRISPR-Cas13d to efficiently target multiple endogenous immunosuppressive genes on demand, allowing us to silence various combinations of multiple immunosuppressive factors in the TME. Intratumoral AAV-mediated administration of MUCIG (AAV-MUCIG) elicits significant anti-tumor activity with several Cas13d gRNA compositions. TME target expression analysis driven optimization led to a simplified off-the-shelf MUCIG targeting a four gene combination (PGGC: Pdl1, Galectin9, Galectin3 and Cd47 ). AAV-PGGC shows significant in vivo efficacy in syngeneic tumor models. Single cell and flow profiling revealed that AAV-PGGC remodeled the TME by increasing CD8 + T cell infiltration and reducing myeloid-derived immunosuppressive cells (MDSCs). MUCIG thus serves as a universal method to silence multiple immune genes in vivo, and can be delivered via AAV as a therapeutic approach.
Collapse
|
215
|
Zou W, Huang R, Li P, Liu X, Huang Q, Yue J, Liu C. Clinical significance of immune checkpoint proteins in HPV-infected cervical cancer. J Infect Public Health 2023; 16:542-550. [PMID: 36801634 DOI: 10.1016/j.jiph.2023.01.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/19/2022] [Accepted: 01/25/2023] [Indexed: 02/01/2023] Open
Abstract
OBJECTIVE To investigate T cell immunoreceptor with Ig and ITIM domain (TIGIT), V-domain Ig suppressor of T cell activation (VISTA), and lymphocyte-activation gene-3 (LAG-3) expression in pathological tissue of human papillomavirus (HPV)-infected cervical cancer (CC) patients and their relationship with patient prognosis. METHODS Clinical data of 175 patients with HPV-infected CC were collected retrospectively. Tumor tissue sections were stained immunohistochemically for TIGIT, VISTA, and LAG-3. The Kaplan-Meier method calculated patient survival. Univariate and multivariate Cox proportional hazards models analyzed all potential risk factors for survival. RESULTS When combined positive score (CPS)= 1 was used as the cut-off value, the Kaplan-Meier survival curve showed that the progression-free survival (PFS) and overall survival (OS) of patients with positive expression of TIGIT and VISTA are shorter (both p < 0.05). Univariate COX regression analysis showed that the positive expression of TIGIT and VISTA are related to patient PFS and OS (both HR>1.0 and p < 0.05). Multivariate COX regression analysis showed that TIGIT-positive patients had shorter OS and VISTA-positive patients had shorter PFS (both HR>1.0 and p < 0.05). There is no significant correlation between LAG-3 expression and PFS or OS. When CPS= 10 was used as the cut-off value, Kaplan-Meier survival curve showed that TIGIT-positive patients had shorter OS (p = 0.019). Univariate COX regression analysis showed that TIGIT-positive expression was associated with the OS of patients (HR=2.209, CI: 1.118-4.365, p = 0.023). However, multivariate COX regression analysis showed that TIGIT expression was not associated significantly with OS. There was no significant correlation between VISTA and LAG-3 expression and PFS or OS. CONCLUSION TIGIT and VISTA are associated closely with HPV-infected CC prognosis and are effective biomarkers.
Collapse
Affiliation(s)
- Wenxue Zou
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| | - Rui Huang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| | - Peihang Li
- Department of Radiation Oncology, Tianjin Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.
| | - Xiang Liu
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Gannan Medical University, Ganzhou Institute and Hospital, Jiangxi, China.
| | - Qingyu Huang
- Department of Graduate, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| | - Jinbo Yue
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| | - Chao Liu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
216
|
Nikoo M, Rabiee F, Mohebbi H, Eghbalifard N, Rajabi H, Yazdani Y, Sakhaei D, Khosravifarsani M, Akhavan-Sigari R. Nivolumab plus ipilimumab combination therapy in cancer: Current evidence to date. Int Immunopharmacol 2023; 117:109881. [PMID: 37012882 DOI: 10.1016/j.intimp.2023.109881] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 01/29/2023] [Accepted: 02/06/2023] [Indexed: 03/06/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized cancer immunotherapy, yielding significant antitumor responses across multiple cancer types. Combination ICI therapy with anti-CTLA-4 and anti-PD-1 antibodies outperforms either antibody alone in terms of clinical efficacy. As a consequence, the U.S. Food and Drug Administration (FDA) approved ipilimumab (anti-CTLA-4) plus nivolumab (anti-PD-1) as the first-ever approved therapies for combined ICI in patients with metastatic melanoma. Despite the success of ICIs, treatment with checkpoint inhibitor combinations poses significant clinical challenges, such as increased rates of immune-related adverse events (irAEs) and drug resistance. Thus, identifying optimal prognostic biomarkers could help to monitor the safety and efficacy of ICIs and identify patients who may benefit the most from these treatments. In this review, we will first go over the fundamentals of the CTLA-4 and PD-1 pathways, as well as the mechanisms of ICI resistance. The results of clinical findings that evaluated the combination of ipilimumab and nivolumab are then summarized to support future research in the field of combination therapy. Finally, the irAEs associated with combined ICI therapy, as well as the underlying biomarkers involved in their management, are discussed.
Collapse
|
217
|
Lodde GC, Jansen P, Herbst R, Terheyden P, Utikal J, Pföhler C, Ulrich J, Kreuter A, Mohr P, Gutzmer R, Meier F, Dippel E, Weichenthal M, Sucker A, Placke JM, Zaremba A, Albrecht LJ, Kowall B, Galetzka W, Becker JC, Tasdogan A, Zimmer L, Livingstone E, Hadaschik E, Schadendorf D, Ugurel S, Griewank K. Characterisation and outcome of RAC1 mutated melanoma. Eur J Cancer 2023; 183:1-10. [PMID: 36773463 DOI: 10.1016/j.ejca.2023.01.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 01/08/2023] [Accepted: 01/09/2023] [Indexed: 01/19/2023]
Abstract
BACKGROUND Activating hot spot R29S mutations in RAC1, a small GTPase influencing several cellular processes including cell proliferation and cytoskeleton rearrangement, have been reported in up to 9% of sun-exposed melanomas. Clinical characteristics and treatment implications of RAC1 mutations in melanoma remain unclear. METHODS We investigated the largest set (n = 64) of RAC1 mutated melanoma patients reported to date, including a retrospective single institution cohort (n = 34) from the University Hospital Essen and a prospective multicentre cohort (n = 30) from the translational study Tissue Registry in Melanoma (TRIM; CA209-578), for patient and tumour characteristics as well as therapy outcomes. RESULTS From 3037 sequenced melanoma samples screened RAC1 mutations occurred in ∼2% of samples (64/3037). The most common RAC1 mutation was P29S (95%, 61/64). The majority of tumours had co-occuring MAP kinase mutations (88%, 56/64); mostly activating NRAS (47%, 30/64) mutations, followed by activating BRAF (28%, 18/64) and NF1 (25%, 16/64) mutations. RAC1 mutated melanomas were almost exclusively of cutaneous origin (84%, 54/64) or of unknown primary (MUP, 14%, 9/64). C > T alterations were the most frequent mutation type identified demonstrating a UV-signature for RAC1 mutated melanoma. Most patients with unresectable disease (39) received immune checkpoint inhibitors (ICI) (77%, 30/39). Objective response rate of first-line treatment in patients with stage III/IV disease was 21%; median overall survival was 47.8 months. CONCLUSIONS RAC1 mutated melanomas are rare, mostly of cutaneous origin and frequently harbour concomitant MAP kinase mutations, particularly in NRAS. Patients with advanced disease benefit from systemic treatment with ICI.
Collapse
Affiliation(s)
- Georg C Lodde
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, Essen, Germany.
| | - Philipp Jansen
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, Essen, Germany; Department of Dermatology and Allergology, UK Bonn, Bonn, Germany.
| | - Rudolf Herbst
- Department of Dermatology, Helios Klinikum Erfurt, Erfurt, Germany.
| | | | - Jochen Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany, Department of Dermatology, Venereology and Allergology, Ruprecht-Karl University of Heidelberg, Mannheim, Germany, DFKZ-Hector Cancer Institute, University Medical Center Mannheim, Mannheim, Germany; German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Claudia Pföhler
- Saarland University Medical School, Homburg, Department of Dermatology, Homburg/Saar, Germany.
| | - Jens Ulrich
- Department of Dermatology and Skin Cancer Center, Harzklinikum Dorothea Christiane Erxleben, Quedlinburg, Germany.
| | - Alexander Kreuter
- Department of Dermatology, Venereology and Allergology, HELIOS St. Elisabeth Klinik Oberhausen, University Witten/Herdecke, Oberhausen, Germany.
| | - Peter Mohr
- Dermatological Center Buxtehude, Elbe Kliniken Buxtehude, Buxtehude, Germany.
| | - Ralf Gutzmer
- Department of Dermatology, Venereology, Allergology and Phlebology, University Hospital Mühlenkreiskliniken Minden, Minden, Germany.
| | - Friedegund Meier
- Skin Cancer Center at the University Cancer Centre Dresden and National Center for Tumor Diseases, Dresden, Department of Dermatology, University Hospital Carl Gustav Carus, TU Dresden, Germany, National Center for Tumor Diseases Dresden (NCT/UCC), Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany; German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Edgar Dippel
- Department of Dermatology, Ludwigshafen Medical Center, Ludwigshafen, Germany.
| | - Michael Weichenthal
- Department of Dermatology, Skin Cancer Center, Schleswig-Holstein University Hospital, Campus Kiel, Kiel, Germany.
| | - Antje Sucker
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, Essen, Germany.
| | - Jan-Malte Placke
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, Essen, Germany.
| | - Anne Zaremba
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, Essen, Germany.
| | - Lea Jessica Albrecht
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, Essen, Germany.
| | - Bernd Kowall
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital Essen, Essen, Germany.
| | - Wolfgang Galetzka
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital Essen, Essen, Germany.
| | - Jürgen C Becker
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, Essen, Germany; Translational Skin Cancer Research, University Medicine Essen, Essen, Germany; German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Alpaslan Tasdogan
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, Essen, Germany.
| | - Lisa Zimmer
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, Essen, Germany.
| | - Elisabeth Livingstone
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, Essen, Germany.
| | - Eva Hadaschik
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, Essen, Germany.
| | - Dirk Schadendorf
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, Essen, Germany; German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Selma Ugurel
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, Essen, Germany; German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Klaus Griewank
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, Essen, Germany.
| |
Collapse
|
218
|
Jung J, Heo YJ, Park S. High tumor mutational burden predicts favorable response to anti-PD-(L)1 therapy in patients with solid tumor: a real-world pan-tumor analysis. J Immunother Cancer 2023; 11:jitc-2022-006454. [PMID: 37094985 PMCID: PMC10152061 DOI: 10.1136/jitc-2022-006454] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/02/2023] [Indexed: 04/26/2023] Open
Abstract
BACKGROUND Tumor mutation burden (TMB) is an important biomarker to predict response to anti-PD-L1 treatment across cancer types. TruSight Oncology 500 (TSO500) is currently used globally as a routine assay for TMB. METHODS Between 2019 and 2021, 1744 patients with cancer received TSO500 assay as part of a real-world clinical practice at the Samsung Medical Center, and 426 received anti-PD-(L)1 treatment. Correlations between TMB and clinical outcomes of anti-PD-(L)1 were analyzed. Digital spatial profiling (DSP) was used to investigate the tumor immune environment's influence on the treatment response to anti-PD-(L)1 in high TMB (TMB-H) patients (n=8). RESULTS The incidence of TMB-H (≥10 mutations (mt)/megabase (Mb)) was 14.7% (n=257). Among TMB-H patients, the most common cancer type was colorectal cancer (n=108, 42.0%), followed by gastric cancer (GC; n=49, 19.1%), bladder cancer (n=21, 8.2%), cholangiocarcinoma (n=21, 8.2%), non-small cell lung cancer (n=17, 6.6%), melanoma (n=8, 3.1%), gallbladder cancer (GBC; n=7, 2.7%), and others (n=26, 10.1%). The response rate to anti-PD-(L)1 therapy was substantially higher in GC (71.4% vs 25.8%), GBC (50.0% vs 12.5%), head and neck cancer (50.0% vs 11.1%), and melanoma (71.4% vs 50.7%) among TMB-H patients when compared with low TMB (TMB-L) (<10 mt/Mb) patients with statistical significance. Additional analysis of patients with TMB ≥16 mt/Mb demonstrated prolonged survival after anti-PD-(L)1 therapy compared with patients with TMB-L (not reached vs 418 days, p=0.03). The benefit of TMB ≥16 mt/Mb was greater when combined with microsatellite status and PD-L1 expression profiles. Among the TMB-H patients, those who responded to anti-PD-L1 therapy had numerous active immune cells that infiltrated the tumor regions during the DSP analysis. Natural killer cells (p=0.04), cytotoxic T cells (p<0.01), memory T cells (p<0.01), naïve memory T cells (p<0.01), and proteins related to T-cell proliferation (p<0.01) were observed in a responder group compared with a non-responder group. In contrast, exhausted T-cell and M2 macrophage counts were increased in the non-responder group. CONCLUSIONS The overall incidence of TMB status was analyzed by the TSO500 assay, and TMB-H was observed in 14.7% of the pan-cancer population. In a real-world setting, TMB-H identified by a target sequencing panel seemed to predict response to anti-PD-(L)1 therapy, especially in patients with a higher proportion of immune cells enriched in the tumor region.
Collapse
Affiliation(s)
- Jaeyun Jung
- Innovative Institute for Precision Medicine, Samsung Medical Center, Seoul, Korea (the Republic of)
| | - You Jeong Heo
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea, Seoul, Gangnam-gu, Korea (the Republic of)
| | - Sehhoon Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea, Seoul, Gangnam-gu, Korea (the Republic of)
| |
Collapse
|
219
|
Zhang J, Tu H, Zheng Z, Zhao X, Lin X. RNF31 promotes tumorigenesis via inhibiting RIPK1 kinase-dependent apoptosis. Oncogene 2023; 42:1585-1596. [PMID: 36997719 DOI: 10.1038/s41388-023-02669-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 03/06/2023] [Accepted: 03/13/2023] [Indexed: 04/01/2023]
Abstract
It is well established that interferon (IFN) and tumor necrosis factor (TNF) could synergistically promote antitumor toxicity and avoid resistance of antigen-negative tumors during cancer immunotherapy. The linear ubiquitin chain assembly complex (LUBAC) has been widely known to regulate receptor-interacting protein kinase-1(RIPK1) kinase activity and TNF-mediated cell death during inflammation and embryogenesis. However, whether LUBAC and RIPK1 kinase activity in tumor microenvironment could regulate antitumor immunity are still not very clear. Here, we demonstrated a cancer cell-intrinsic role of LUBAC complex in tumor microenvironment to promote tumorigenesis. Lacking LUBAC component RNF31 in B16 melanoma cells but not immune cells including macrophages or dendritic cells greatly impaired tumor growth by increasing intratumoral CD8+ T cells infiltration. Mechanistically, we found that tumor cells without RNF31 shown severe apoptosis-mediated cell death caused by TNFα/IFNγ in the tumor microenvironment. Most importantly, we found that RNF31 could limit RIPK1 kinase activity and further prevent tumor cell death in a transcription-independent manner, suggesting a crucial role of RIPK1 kinase activity in tumorigenesis. Together, our results demonstrate an essential role of RNF31 and RIPK1 kinase activity in tumorigenesis and imply that RNF31 inhibition could be harnessed to enhance antitumor toxicity during tumor immunotherapy.
Collapse
Affiliation(s)
- Jie Zhang
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China
- Department of Basic Medical Sciences, Tsinghua University School of Medicine, Beijing, 100084, China
| | - Hailin Tu
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China
- Department of Basic Medical Sciences, Tsinghua University School of Medicine, Beijing, 100084, China
| | - Zheyu Zheng
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China
- Department of Basic Medical Sciences, Tsinghua University School of Medicine, Beijing, 100084, China
| | - Xueqiang Zhao
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China
- Department of Basic Medical Sciences, Tsinghua University School of Medicine, Beijing, 100084, China
| | - Xin Lin
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China.
- Department of Basic Medical Sciences, Tsinghua University School of Medicine, Beijing, 100084, China.
| |
Collapse
|
220
|
Walsh MJ, Stump CT, Kureshi R, Lenehan P, Ali LR, Dougan M, Knipe DM, Dougan SK. IFNγ is a central node of cancer immune equilibrium. Cell Rep 2023; 42:112219. [PMID: 36881506 PMCID: PMC10214249 DOI: 10.1016/j.celrep.2023.112219] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 01/09/2023] [Accepted: 02/20/2023] [Indexed: 03/08/2023] Open
Abstract
Tumors in immune equilibrium are held in balance between outgrowth and destruction by the immune system. The equilibrium phase defines the duration of clinical remission and stable disease, and escape from equilibrium remains a major clinical problem. Using a non-replicating HSV-1 vector expressing interleukin-12 (d106S-IL12), we developed a mouse model of therapy-induced immune equilibrium, a phenomenon previously seen only in humans. This immune equilibrium was centrally reliant on interferon-γ (IFNγ). CD8+ T cell direct recognition of MHC class I, perforin/granzyme-mediated cytotoxicity, and extrinsic death receptor signaling such as Fas/FasL were all individually dispensable for equilibrium. IFNγ was critically important and played redundant roles in host and tumor cells such that IFNγ sensing in either compartment was sufficient for immune equilibrium. We propose that these redundant mechanisms of action are integrated by IFNγ to protect from oncogenic or chronic viral threats and establish IFNγ as a central node in therapy-induced immune equilibrium.
Collapse
Affiliation(s)
- Michael J Walsh
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Harvard Program in Virology, Boston, MA, USA; Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Courtney T Stump
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Rakeeb Kureshi
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Patrick Lenehan
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Lestat R Ali
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Michael Dougan
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - David M Knipe
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA.
| | - Stephanie K Dougan
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Immunology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
221
|
Shi MW, Huang J, Sun Y. Neoadjuvant Immunotherapy for Head and Neck Squamous Cell Carcinoma: Expecting Its Application in Temporal Bone Squamous Cell Carcinoma. Curr Med Sci 2023; 43:213-222. [PMID: 36971976 DOI: 10.1007/s11596-023-2700-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 09/14/2022] [Indexed: 03/29/2023]
Abstract
Temporal bone malignant tumors are characterized by atypical clinical symptoms, and easy recurrence and metastasis. They account for 0.2% of head and neck tumors, and the most common pathological type is squamous cell carcinoma. Patients with squamous cell carcinoma of the temporal bone are often at advanced stages when diagnosed, and lose the chance for surgery. Neoadjuvant immunotherapy has recently been approved as the first-line treatment for refractory recurrent/metastatic squamous cell carcinoma of the head and neck. However, it remains to be determined whether neoadjuvant immunotherapy can be used as the first-line treatment for temporal bone squamous cell carcinoma to reduce the tumor stage before surgery, or as a palliative treatment for patients with unresectable advanced stage carcinoma. The present study reviews the development of immunotherapy and its clinical application in head and neck squamous cell carcinoma, summarizes the treatment of temporal bone squamous cell carcinoma, and prospects the neoadjuvant immunotherapy as the first-line treatment for temporal bone squamous cell carcinoma.
Collapse
Affiliation(s)
- Meng-Wen Shi
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jing Huang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Yu Sun
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
222
|
Regal JA, Guerra García ME, Jain V, Chandramohan V, Ashley DM, Gregory SG, Thompson EM, López GY, Reitman ZJ. Ganglioglioma deep transcriptomics reveals primitive neuroectoderm neural precursor-like population. Acta Neuropathol Commun 2023; 11:50. [PMID: 36966348 PMCID: PMC10039537 DOI: 10.1186/s40478-023-01548-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 03/06/2023] [Indexed: 03/27/2023] Open
Abstract
Gangliogliomas are brain tumors composed of neuron-like and macroglia-like components that occur in children and young adults. Gangliogliomas are often characterized by a rare population of immature astrocyte-appearing cells expressing CD34, a marker expressed in the neuroectoderm (neural precursor cells) during embryogenesis. New insights are needed to refine tumor classification and to identify therapeutic approaches. We evaluated five gangliogliomas with single nucleus RNA-seq, cellular indexing of transcriptomes and epitopes by sequencing, and/or spatially-resolved RNA-seq. We uncovered a population of CD34+ neoplastic cells with mixed neuroectodermal, immature astrocyte, and neuronal markers. Gene regulatory network interrogation in these neuroectoderm-like cells revealed control of transcriptional programming by TCF7L2/MEIS1-PAX6 and SOX2, similar to that found during neuroectodermal/neural development. Developmental trajectory analyses place neuroectoderm-like tumor cells as precursor cells that give rise to neuron-like and macroglia-like neoplastic cells. Spatially-resolved transcriptomics revealed a neuroectoderm-like tumor cell niche with relative lack of vascular and immune cells. We used these high resolution results to deconvolute clinically-annotated transcriptomic data, confirming that CD34+ cell-associated gene programs associate with gangliogliomas compared to other glial brain tumors. Together, these deep transcriptomic approaches characterized a ganglioglioma cellular hierarchy-confirming CD34+ neuroectoderm-like tumor precursor cells, controlling transcription programs, cell signaling, and associated immune cell states. These findings may guide tumor classification, diagnosis, prognostication, and therapeutic investigations.
Collapse
Affiliation(s)
- Joshua A Regal
- Department of Radiation Oncology, Duke University, Durham, NC, 27710, USA
| | | | - Vaibhav Jain
- Duke Molecular Physiology Institute, Duke University, Durham, NC, 27710, USA
| | | | - David M Ashley
- Department of Neurosurgery, Duke University, Durham, NC, 27710, USA
| | - Simon G Gregory
- Duke Molecular Physiology Institute, Duke University, Durham, NC, 27710, USA
| | - Eric M Thompson
- Department of Neurosurgery, Duke University, Durham, NC, 27710, USA
| | - Giselle Y López
- Department of Neurosurgery, Duke University, Durham, NC, 27710, USA
- Department of Pathology, Duke University, Durham, NC, 27710, USA
| | - Zachary J Reitman
- Department of Radiation Oncology, Duke University, Durham, NC, 27710, USA.
- Department of Neurosurgery, Duke University, Durham, NC, 27710, USA.
- Department of Pathology, Duke University, Durham, NC, 27710, USA.
| |
Collapse
|
223
|
Diaz MJ, Mark I, Rodriguez D, Gelman B, Tran JT, Kleinberg G, Levin A, Beneke A, Root KT, Tran AXV, Lucke-Wold B. Melanoma Brain Metastases: A Systematic Review of Opportunities for Earlier Detection, Diagnosis, and Treatment. Life (Basel) 2023; 13:life13030828. [PMID: 36983983 PMCID: PMC10053844 DOI: 10.3390/life13030828] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 02/25/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Introduction: Melanoma continues to represent the most serious skin cancer worldwide. However, few attempts have been made to connect the body of research on advanced melanoma. In the present review, we report on strides made in the diagnosis and treatment of intracranial metastatic melanoma. Methods: Relevant Cochrane reviews and randomized-controlled trials published by November 2022 were systematically retrieved from the Cochrane Library, EMBASE, and PubMed databases (N = 27). Search and screening methods adhered to the 2020 revision of the Preferred Reporting Items for Systematic reviews and Meta-Analyses guidelines. Results: Although the research surrounding the earlier detection of melanoma brain metastasis is scarce, several studies have highlighted specific markers associated with MBM. Such factors include elevated BRAFV600 mutant ctDNA, high LDH concentration, and high IGF-1R. The approach to treating MBM is moving away from surgery and toward nonsurgical management, namely, a combination of stereotactic radiosurgery (SRS) and immunotherapeutic agents. There is an abundance of emerging research seeking to identify and improve both novel and established treatment options and diagnostic approaches for MBM, however, more research is still needed to maximize the clinical efficacy, especially for new immunotherapeutics. Conclusions: Early detection is optimal for the efficacy of treatment and MBM prognosis. Current treatment utilizes chemotherapies and targeted therapies. Emerging approaches emphasize biomarkers and joint treatments. Further exploration toward preliminary identification, the timing of therapies, and methods to ameliorate adverse treatment effects are needed to advance MBM patient care.
Collapse
Affiliation(s)
| | - Isabella Mark
- College of Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Daphnee Rodriguez
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Beata Gelman
- Department of Biology, University of Maryland, College Park, MD 20742, USA
| | - Jasmine Thuy Tran
- School of Medicine, University of Indiana, Indianapolis, IN 46202, USA
| | - Giona Kleinberg
- College of Engineering, Northeastern University, Boston, MA 02115, USA
| | - Anna Levin
- School of Arts and Sciences, Rutgers University, Piscataway, NJ 08854, USA
| | - Alice Beneke
- College of Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Kevin Thomas Root
- College of Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Andrew Xuan Vinh Tran
- Department of Dermatology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
224
|
Zhao Z, Huang Y, Wang J, Lin H, Cao F, Li S, Li Y, Li Z, Liu X. A self-assembling CXCR4-targeted pyroptosis nanotoxin for melanoma therapy. Biomater Sci 2023; 11:2200-2210. [PMID: 36745434 DOI: 10.1039/d2bm02026b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
While immunotherapy has emerged as a promising strategy to treat melanoma, the limited availability of immunotherapeutic agents in tumors due to the immunosuppressive tumor microenvironment dampens its efficacy. Pyroptosis is a gasdermin-mediated programmed necrosis that triggers the inflammatory tumor microenvironment and enhances the efficacy of tumor immunotherapy. Here, we prove that the CXCR4 antagonist T22 peptide specially targeted and became internalized into CXCR4+ melanoma cells. Then we report a self-assembling nanotoxin that can be used to spatiotemporally target CXCR4-expression melanoma cells and enable tunable cellular pyroptosis. Specific activation of caspase 3 signal transduction triggers gasdermin-E-mediated pyroptosis. This nanotoxin induces pyroptotic cell death resulting in enhanced antitumor efficacy and minimized systemic side effects toward melanoma in vivo. This study offers new insights into how to engineer nanotoxins with tunable pyroptosis activity through specifically targeting CXCR4 for biomedical applications.
Collapse
Affiliation(s)
- Zheng Zhao
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China.
| | - Yingbin Huang
- Organ Transplantation Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jing Wang
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China.
| | - Hongsheng Lin
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China.
| | - Fei Cao
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China.
| | - Shuxin Li
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China.
| | - Yin Li
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China.
| | - Ziqian Li
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China.
| | - Xuekui Liu
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China.
| |
Collapse
|
225
|
Luo Y, Ye Y, Chen Y, Zhang C, Sun Y, Wang C, Ou J. A degradome-based prognostic signature that correlates with immune infiltration and tumor mutation burden in breast cancer. Front Immunol 2023; 14:1140993. [PMID: 36993976 PMCID: PMC10040797 DOI: 10.3389/fimmu.2023.1140993] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 02/27/2023] [Indexed: 03/14/2023] Open
Abstract
IntroductionFemale breast cancer is the most common malignancy worldwide, with a high disease burden. The degradome is the most abundant class of cellular enzymes that play an essential role in regulating cellular activity. Dysregulation of the degradome may disrupt cellular homeostasis and trigger carcinogenesis. Thus we attempted to understand the prognostic role of degradome in breast cancer by means of establishing a prognostic signature based on degradome-related genes (DRGs) and assessed its clinical utility in multiple dimensions.MethodsA total of 625 DRGs were obtained for analysis. Transcriptome data and clinical information of patients with breast cancer from TCGA-BRCA, METABRIC and GSE96058 were collected. NetworkAnalyst and cBioPortal were also utilized for analysis. LASSO regression analysis was employed to construct the degradome signature. Investigations of the degradome signature concerning clinical association, functional characterization, mutation landscape, immune infiltration, immune checkpoint expression and drug priority were orchestrated. Cell phenotype assays including colony formation, CCK8, transwell and wound healing were conducted in MCF-7 and MDA-MB-435S breast cancer cell lines, respectively.ResultsA 10-gene signature was developed and verified as an independent prognostic predictor combined with other clinicopathological parameters in breast cancer. The prognostic nomogram based on risk score (calculated based on the degradome signature) showed favourable capability in survival prediction and advantage in clinical benefit. High risk scores were associated with a higher degree of clinicopathological events (T4 stage and HER2-positive) and mutation frequency. Regulation of toll-like receptors and several cell cycle promoting activities were upregulated in the high-risk group. PIK3CA and TP53 mutations were dominant in the low- and high-risk groups, respectively. A significantly positive correlation was observed between the risk score and tumor mutation burden. The infiltration levels of immune cells and the expressions of immune checkpoints were significantly influenced by the risk score. Additionally, the degradome signature adequately predicted the survival of patients undergoing endocrinotherapy or radiotherapy. Patients in the low-risk group may achieve complete response after the first round of chemotherapy with cyclophosphamide and docetaxel, whereas patients in the high-risk group may benefit from 5-flfluorouracil. Several regulators of the PI3K/AKT/mTOR signaling pathway and the CDK family/PARP family were identified as potential molecular targets in the low- and high-risk groups, respectively. In vitro experiments further revealed that the knockdown of ABHD12 and USP41 significantly inhibit the proliferation, invasion and migration of breast cancer cells.ConclusionMultidimensional evaluation verified the clinical utility of the degradome signature in predicting prognosis, risk stratification and guiding treatment for patients with breast cancer.
Collapse
Affiliation(s)
- Yulou Luo
- Department of Breast Surgery, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, China
| | - Yinghui Ye
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yan Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, China
| | - Chenguang Zhang
- Department of Breast Surgery, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, China
| | - Yutian Sun
- Department of Medical Oncology, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Chengwei Wang
- Cancer Research Institute of Xinjiang Uygur Autonomous Region, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, China
- *Correspondence: Chengwei Wang, ; Jianghua Ou,
| | - Jianghua Ou
- Department of Breast Surgery, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, China
- *Correspondence: Chengwei Wang, ; Jianghua Ou,
| |
Collapse
|
226
|
Li S, Zou D, Liu Z. Comprehensive bioinformatic analysis constructs a CXCL model for predicting survival and immunotherapy effectiveness in ovarian cancer. Front Pharmacol 2023; 14:1127557. [PMID: 36969851 PMCID: PMC10034089 DOI: 10.3389/fphar.2023.1127557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/27/2023] [Indexed: 03/11/2023] Open
Abstract
Background: Immunotherapy has limited effectiveness in ovarian cancer (OC) patients, highlighting the need for reliable biomarkers to predict the effectiveness of these treatments. The C-X-C motif chemokine ligands (CXCLs) have been shown to be associated with survival outcomes and immunotherapy efficacy in cancer patients. In this study, we aimed to evaluate the predictive value of 16 CXCLs in OC patients.Methods: We analyzed RNA-seq data from The Cancer Genome Atlas, Gene Expression Omnibus, and UCSC Xena database and conducted survival analysis. Consensus cluster analysis was used to group patients into distinct clusters based on their expression patterns. Biological pathway alterations and immune infiltration patterns were examined across these clusters using gene set variation analysis and single-sample gene set enrichment analysis. We also developed a CXCL scoring model using principal component analysis and evaluated its effectiveness in predicting immunotherapy response by assessing tumor microenvironment cell infiltration, tumor mutational burden estimation, PD-L1/CTLA4 expression, and immunophenoscore analysis (IPS).Results: Most CXCL family genes were overexpressed in OC tissues compared to normal ovarian tissues. Patients were grouped into three distinct CXCL clusters based on their CXCL expression pattern. Additionally, using differentially expressed genes among the CXCL clusters, patients could also be grouped into three gene clusters. The CXCL and gene subtypes effectively predicted survival and immune cell infiltration levels for OC patients. Furthermore, patients with high CXCL scores had significantly better survival outcomes, higher levels of immune cell infiltration, higher IPS, and higher expression of PD-L1/CTLA4 than those with low CXCL scores.Conclusion: The CXCL score has the potential to be a promising biomarker to guide immunotherapy in individual OC patients and predict their clinical outcomes and immunotherapy responses.
Collapse
Affiliation(s)
- Shuang Li
- Hunan Key Laboratory of Pharmacogenetics, Department of Clinical Pharmacology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Dawei Zou
- Department of Surgery, Immunobiology and Transplant Science Center, Houston Methodist Research Institute and Institute for Academic Medicine, Houston Methodist Hospital, Houston, TX, United States
- *Correspondence: Zhaoqian Liu, ; Dawei Zou,
| | - Zhaoqian Liu
- Hunan Key Laboratory of Pharmacogenetics, Department of Clinical Pharmacology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacology, Central South University, Changsha, China
- *Correspondence: Zhaoqian Liu, ; Dawei Zou,
| |
Collapse
|
227
|
Burtness B. First-Line Nivolumab Plus Ipilimumab in Recurrent/Metastatic Head and Neck Cancer-What Happened? J Clin Oncol 2023; 41:2134-2137. [PMID: 36877893 DOI: 10.1200/jco.22.02349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023] Open
Affiliation(s)
- Barbara Burtness
- Department of Internal Medicine, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| |
Collapse
|
228
|
Zhou X, Fu C, Chen X. The role of ubiquitin pathway-mediated regulation of immune checkpoints in cancer immunotherapy. Cancer 2023; 129:1649-1661. [PMID: 36857206 DOI: 10.1002/cncr.34729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 02/09/2023] [Accepted: 02/14/2023] [Indexed: 03/02/2023]
Abstract
With the continuous cognition of the relationship between tumor cells and tumor immune microenvironment, immunotherapy based on the immune checkpoint blockade has achieved great breakthroughs, led to improved clinical outcomes, and prolonged survival for cancer patients in recent years. Nevertheless, the de novo or acquired resistance to immunotherapy has greatly counteracted the efficacy, leading to a 20%-40% overall response rate. Thus, further in-depth understanding of the regulation of the tumor microenvironment and antitumor immunity is urgently warranted. Ubiquitination-mediated protein degradation plays vital roles in protein stabilization, activation, and dynamics as well as in cellular homeostasis modulation. The dysregulated ubiquitination and deubiquitination are closely related to the changes in physiological and pathological processes, which subsequently result in a variety of diseases including cancer. In this review, the authors first summarize the current knowledge about the involvement of the ubiquitin-proteasome system in tumor development with the ubiquitin conjugation-regulated stability of p53, phosphatase and tensin homolog, and Myc protein as examples, then dissect the potential implications of ubiquitination-mediated immune checkpoints degradation in tumor microenvironment and immune responses, and finally discuss the effects of therapeutically targeting the ubiquitin-proteasome pathway on immunotherapy, with the goal of providing deep insights into the exploitation of more precise and effective combinational therapy against cancer.
Collapse
Affiliation(s)
- Xiaoming Zhou
- Cancer Research Institute, the First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China.,Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Chengxiao Fu
- Cancer Research Institute, the First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China.,Department of Pharmacy, the First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xisha Chen
- Cancer Research Institute, the First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China.,Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
229
|
Edwards-Hicks J, Apostolova P, Buescher JM, Maib H, Stanczak MA, Corrado M, Klein Geltink RI, Maccari ME, Villa M, Carrizo GE, Sanin DE, Baixauli F, Kelly B, Curtis JD, Haessler F, Patterson A, Field CS, Caputa G, Kyle RL, Soballa M, Cha M, Paul H, Martin J, Grzes KM, Flachsmann L, Mitterer M, Zhao L, Winkler F, Rafei-Shamsabadi DA, Meiss F, Bengsch B, Zeiser R, Puleston DJ, O'Sullivan D, Pearce EJ, Pearce EL. Phosphoinositide acyl chain saturation drives CD8 + effector T cell signaling and function. Nat Immunol 2023; 24:516-530. [PMID: 36732424 PMCID: PMC10908374 DOI: 10.1038/s41590-023-01419-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 01/03/2023] [Indexed: 02/04/2023]
Abstract
How lipidome changes support CD8+ effector T (Teff) cell differentiation is not well understood. Here we show that, although naive T cells are rich in polyunsaturated phosphoinositides (PIPn with 3-4 double bonds), Teff cells have unique PIPn marked by saturated fatty acyl chains (0-2 double bonds). PIPn are precursors for second messengers. Polyunsaturated phosphatidylinositol bisphosphate (PIP2) exclusively supported signaling immediately upon T cell antigen receptor activation. In late Teff cells, activity of phospholipase C-γ1, the enzyme that cleaves PIP2 into downstream mediators, waned, and saturated PIPn became essential for sustained signaling. Saturated PIP was more rapidly converted to PIP2 with subsequent recruitment of phospholipase C-γ1, and loss of saturated PIPn impaired Teff cell fitness and function, even in cells with abundant polyunsaturated PIPn. Glucose was the substrate for de novo PIPn synthesis, and was rapidly utilized for saturated PIP2 generation. Thus, separate PIPn pools with distinct acyl chain compositions and metabolic dependencies drive important signaling events to initiate and then sustain effector function during CD8+ T cell differentiation.
Collapse
Affiliation(s)
- Joy Edwards-Hicks
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Petya Apostolova
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Joerg M Buescher
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Hannes Maib
- Division of Cell & Developmental Biology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Michal A Stanczak
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mauro Corrado
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | | | - Maria Elena Maccari
- Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Matteo Villa
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Gustavo E Carrizo
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - David E Sanin
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Francesc Baixauli
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Beth Kelly
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jonathan D Curtis
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Fabian Haessler
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Annette Patterson
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Cameron S Field
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - George Caputa
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Ryan L Kyle
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Melanie Soballa
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Minsun Cha
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Harry Paul
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jacob Martin
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Katarzyna M Grzes
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lea Flachsmann
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Michael Mitterer
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Liang Zhao
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Frances Winkler
- Clinic for Internal Medicine II, Gastroenterology, Hepatology, Endocrinology, and Infectious Diseases, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - David Ali Rafei-Shamsabadi
- Department of Dermatology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Frank Meiss
- Department of Dermatology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Bertram Bengsch
- Clinic for Internal Medicine II, Gastroenterology, Hepatology, Endocrinology, and Infectious Diseases, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Signaling Research Centers BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Robert Zeiser
- Signaling Research Centers BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Daniel J Puleston
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - David O'Sullivan
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Edward J Pearce
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Erika L Pearce
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany.
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
230
|
Cai Z, Chen J, Yu Z, Li H, Liu Z, Deng D, Liu J, Chen C, Zhang C, Ou Z, Chen M, Hu J, Zu X. BCAT2 Shapes a Noninflamed Tumor Microenvironment and Induces Resistance to Anti-PD-1/PD-L1 Immunotherapy by Negatively Regulating Proinflammatory Chemokines and Anticancer Immunity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207155. [PMID: 36642843 PMCID: PMC10015882 DOI: 10.1002/advs.202207155] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Indexed: 06/17/2023]
Abstract
To improve response rate of monotherapy of immune checkpoint blockade (ICB), it is necessary to find an emerging target in combination therapy. Through analyzing tumor microenvironment (TME)-related indicators, it is validated that BCAT2 shapes a noninflamed TME in bladder cancer. The outcomes of multiomics indicate that BCAT2 has an inhibitory effect on cytotoxic lymphocyte recruitment by restraining activities of proinflammatory cytokine/chemokine-related pathways and T-cell-chemotaxis pathway. Immunoassays reveal that secretion of CD8+ T-cell-related chemokines keeps a robust negative correlation with BCAT2, generating a decreasing tendency of CD8+ T cells around BCAT2+ tumor cells from far to near. Cotreatment of BCAT2 deficiency and anti-PD-1 antibody has a synergistic effect in vivo, implying the potential of BCAT2 in combination therapy. Moreover, the value of BCAT2 in predicting efficacy of immunotherapy is validated in multiple immunotherapy cohorts. Together, as a key molecule in TME, BCAT2 is an emerging target in combination with ICB and a biomarker of guiding precision therapy.
Collapse
Affiliation(s)
- Zhiyong Cai
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Jinbo Chen
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Zhengzheng Yu
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- Research Center of Carcinogenesis and Targeted TherapyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Huihuang Li
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Zhi Liu
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Dingshan Deng
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Jinhui Liu
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Chunliang Chen
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Chunyu Zhang
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Zhenyu Ou
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Minfeng Chen
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Jiao Hu
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Xiongbing Zu
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| |
Collapse
|
231
|
Liu M, Wang X, Du X, Wu W, Zhang Y, Zhang P, Ai C, Devenport M, Su J, Muthana MM, Su L, Liu Y, Zheng P. Soluble CTLA-4 mutants ameliorate immune-related adverse events but preserve efficacy of CTLA-4- and PD-1-targeted immunotherapy. Sci Transl Med 2023; 15:eabm5663. [PMID: 36857433 PMCID: PMC10501849 DOI: 10.1126/scitranslmed.abm5663] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 01/31/2023] [Indexed: 03/03/2023]
Abstract
Immune checkpoint inhibitors (ICIs), such as nivolumab and ipilimumab, not only elicit antitumor responses in a wide range of human cancers but also cause severe immune-related adverse events (irAEs), including death. A largely unmet medical need is to treat irAEs without abrogating the immunotherapeutic effect of ICIs. Although abatacept has been used to treat irAEs, it risks neutralizing the anti-cytotoxic T lymphocyte-associated protein 4 (CTLA-4) monoclonal antibodies administered for cancer therapy, thereby reducing the efficacy of anti-CTLA-4 immunotherapy. To avoid this caveat, we compared wild-type abatacept and mutants of CTLA-4-Ig for their binding to clinically approved anti-CTLA-4 antibodies and for their effect on both irAEs and immunotherapy conferred by anti-CTLA-4 and anti-PD-1 antibodies. Here, we report that whereas abatacept neutralized the therapeutic effect of anti-CTLA-4 antibodies, the mutants that bound to B7-1 and B7-2, but not to clinical anti-CTLA-4 antibodies, including clinically used belatacept, abrogated irAEs without affecting cancer immunotherapy. Our data demonstrate that anti-CTLA-4-induced irAEs can be corrected by provision of soluble CTLA-4 variants and that the clinically available belatacept may emerge as a broadly applicable drug to abrogate irAEs while preserving the therapeutic efficacy of CTLA-4-targeting ICIs.
Collapse
Affiliation(s)
- Mingyue Liu
- Division of Immunotherapy, Institute of Human Virology and Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Xu Wang
- Division of Immunotherapy, Institute of Human Virology and Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Xuexiang Du
- Division of Immunotherapy, Institute of Human Virology and Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Key Laboratory of Infection and Immunity of Shandong Province & Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
| | - Wei Wu
- Division of Immunotherapy, Institute of Human Virology and Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- OncoC4 Inc., Rockville, MD 20805, USA
| | - Yan Zhang
- Division of Immunotherapy, Institute of Human Virology and Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Immunology and Microbiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Peng Zhang
- Division of Immunotherapy, Institute of Human Virology and Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing 100045, China
| | - Chunxia Ai
- Division of Immunotherapy, Institute of Human Virology and Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Key Laboratory of Infection and Immunity of Shandong Province & Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
| | | | - Juanjuan Su
- Division of Immunotherapy, Institute of Human Virology and Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- OncoC4 Inc., Rockville, MD 20805, USA
| | - Musleh M. Muthana
- Division of Immunotherapy, Institute of Human Virology and Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Lishan Su
- Division of Immunotherapy, Institute of Human Virology and Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Laboratory of Viral Pathogenesis and Immunotherapy, Divisions of Virology, Pathogenesis and Cancer and Immunotherapy, Institute of Human Virology and Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Yang Liu
- Division of Immunotherapy, Institute of Human Virology and Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- OncoC4 Inc., Rockville, MD 20805, USA
| | - Pan Zheng
- Division of Immunotherapy, Institute of Human Virology and Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- OncoC4 Inc., Rockville, MD 20805, USA
| |
Collapse
|
232
|
Henson CE, Morton DJ, Mayadev JS, Wong SJ, Zamarin D. Cancer trials as opportunities to serve and learn from individuals with human immunodeficiency virus. Cancer 2023; 129:664-667. [PMID: 36576320 PMCID: PMC10427029 DOI: 10.1002/cncr.34598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Christina E. Henson
- Department of Radiation Oncology, Stephenson Cancer Center and University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Daniel J. Morton
- Stephenson Cancer Center, Oklahoma City, Oklahoma, USA
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Jyoti S. Mayadev
- Department of Radiation Medicine and Applied Sciences, University of California San Diego School of Medicine, San Diego, California, USA
| | - Stuart J. Wong
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Dmitriy Zamarin
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York, USA
| |
Collapse
|
233
|
Incorvaia L, Rinaldi G, Badalamenti G, Cucinella A, Brando C, Madonia G, Fiorino A, Pipitone A, Perez A, Li Pomi F, Galvano A, Gristina V, Barraco N, Bono M, Bazan Russo TD, Toia F, Cordova A, Fanale D, Russo A, Bazan V. Prognostic role of soluble PD-1 and BTN2A1 in overweight melanoma patients treated with nivolumab or pembrolizumab: finding the missing links in the symbiotic immune-metabolic interplay. Ther Adv Med Oncol 2023; 15:17588359231151845. [PMID: 36818688 PMCID: PMC9936535 DOI: 10.1177/17588359231151845] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 01/04/2023] [Indexed: 02/18/2023] Open
Abstract
Individual response to immune checkpoint inhibitors (ICIs) is currently unpredictable in patients with melanoma. Recent findings highlight a striking improvement in the clinical outcomes of overweight/obese patients treated with ICIs, which seems driven, at least in part, by programmed cell death protein 1 (PD-1)-mediated T-cell dysfunction. A putative role of butyrophilins (BTNs) is under investigation as a novel mechanism of cancer immune evasion and obesity-associated inflammation. This study investigates the role of baseline plasma levels of soluble PD-1 (sPD-1), soluble programmed cell death ligand 1 (sPD-L1), BTN2A1 (sBTN2A1), BTN3A1 (sBTN3A1), along with body mass index (BMI), as predictive biomarkers of immunotherapy response in metastatic melanoma patients treated with nivolumab or pembrolizumab as first-line treatment. In all, 41 patients were included in the study. The baseline plasma level of sPD-1 was significantly lower, and the sBTN2A1 was significantly higher, in long-responder patients to nivolumab or pembrolizumab (median sPD-1: 10.3 ng/ml versus 16.6 ng/ml, p = 0.001; median sBTN2A1: 4.4 ng/ml versus 3.77 ng/ml, p = 0.004). Lower levels of sPD-1 and higher levels of sBTN2A1 were also significantly associated with better overall response rate. Notably, when we further stratified the study cohort using BMI along with sPD-1, patients with BMI ⩾ 25 and sPD-1 < 11.24 ng/ml had longer time to treatment failure after PD-1 inhibitor than other subgroups of patients (p < 0.001). Circulating sPD-1 and sBTN2A1 detection, along with BMI, could give more insights into the immune-metabolic interactions underlying the benefit observed in overweight/obese patients, improving the use of dynamic, noninvasive, biomarkers for patient selection.
Collapse
Affiliation(s)
| | | | | | - Alessandra Cucinella
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Chiara Brando
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Giorgio Madonia
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Alessia Fiorino
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Angela Pipitone
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Alessandro Perez
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Federica Li Pomi
- Department of Clinical and Experimental Medicine, Section of Dermatology, University of Messina, Messina, Italy
| | - Antonio Galvano
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Valerio Gristina
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Nadia Barraco
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Marco Bono
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Tancredi Didier Bazan Russo
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Francesca Toia
- Division of Plastic and Reconstructive Surgery, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Adriana Cordova
- Division of Plastic and Reconstructive Surgery, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Daniele Fanale
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | | | | |
Collapse
|
234
|
Knight A, Karapetyan L, Kirkwood JM. Immunotherapy in Melanoma: Recent Advances and Future Directions. Cancers (Basel) 2023; 15:1106. [PMID: 36831449 PMCID: PMC9954703 DOI: 10.3390/cancers15041106] [Citation(s) in RCA: 89] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/05/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
The use of immunotherapy in the treatment of advanced and high-risk melanoma has led to a striking improvement in outcomes. Although the incidence of melanoma has continued to rise, median survival has improved from approximately 6 months to nearly 6 years for patients with advanced inoperable stage IV disease. Recent understanding of the tumor microenvironment and its interplay with the immune system has led to the explosive development of novel immunotherapy treatments. Since the approval of the therapeutic cytokines interleukin-2 and interferon alfa-2 in the 1990s, the development of novel immune checkpoint inhibitors (ICIs), oncolytic virus therapy, and modulators of the tumor microenvironment have given way to a new era in melanoma treatment. Monoclonal antibodies directed at programmed cell death protein 1 receptor (PD-1) and its ligand (PDL-1), cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), and lymphocyte-activation gene 3 (LAG-3) have provided robust activation of the adaptive immune system, restoring immune surveillance leading to host tumor recognition and destruction. Multiple other immunomodulatory therapeutics are under investigation to overcome resistance to ICI therapy, including the toll-like receptor-9 (TLR-9) and 7/8 (TLR-7/8) agonists, stimulator of interferon genes (STING) agonists, and fecal microbiota transplantation. In this review, we focus on the recent advances in immunotherapy for the treatment of melanoma and provide an update on novel therapies currently under investigation.
Collapse
Affiliation(s)
- Andrew Knight
- Department of Medicine, Division of General Internal Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Lilit Karapetyan
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - John M. Kirkwood
- Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh Medical Center, Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| |
Collapse
|
235
|
Müller-Jensen L, Knauss S, Ginesta Roque L, Schinke C, Maierhof SK, Bartels F, Finke C, Rentzsch K, Ulrich C, Mohr R, Stenzel W, Endres M, Boehmerle W, Huehnchen P. Autoantibody profiles in patients with immune checkpoint inhibitor-induced neurological immune related adverse events. Front Immunol 2023; 14:1108116. [PMID: 36845122 PMCID: PMC9945255 DOI: 10.3389/fimmu.2023.1108116] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 01/23/2023] [Indexed: 02/10/2023] Open
Abstract
Background Neurological immune-related adverse events (irAE-n) are severe and potentially fatal toxicities of immune checkpoint inhibitors (ICI). To date, the clinical significance of neuronal autoantibodies in irAE-n is poorly understood. Here, we characterize neuronal autoantibody profiles in patients with irAE-n and compare these with ICI-treated cancer patients without irAE-n. Methods In this cohort study (DRKS00012668), we consecutively collected clinical data and serum samples of 29 cancer patients with irAE-n (n = 2 pre-ICI, n = 29 post-ICI) and 44 cancer control patients without irAE-n (n = 44 pre- and post-ICI). Using indirect immunofluorescence and immunoblot assays, serum samples were tested for a large panel of neuromuscular and brain-reactive autoantibodies. Results IrAE-n patients and controls received ICI treatment targeting programmed death protein (PD-)1 (61% and 62%), programmed death ligand (PD-L)1 (18% and 33%) or PD-1 and cytotoxic T-lymphocyte-associated protein (CTLA-)4 (21% and 5%). Most common malignancies were melanoma (both 55%) and lung cancer (11% and 14%). IrAE-n affected the peripheral nervous system (59%), the central nervous system (21%), or both (21%). Prevalence of neuromuscular autoantibodies was 63% in irAE-n patients, which was higher compared to ICI-treated cancer patients without irAE-n (7%, p <.0001). Brain-reactive autoantibodies targeting surface (anti-GABABR, -NMDAR, -myelin), intracellular (anti-GFAP, -Zic4, -septin complex), or unknown antigens were detected in 13 irAE-n patients (45%). In contrast, only 9 of 44 controls (20%) presented brain-reactive autoantibodies before ICI administration. However, seven controls developed de novo brain-reactive autoantibodies after ICI initiation, therefore, prevalence of brain-reactive autoantibodies was comparable between ICI-treated patients with and without irAE-n (p = .36). While there was no clear association between specific brain-reactive autoantibodies and clinical presentation, presence of at least one of six selected neuromuscular autoantibodies (anti-titin, anti-skeletal muscle, anti-heart muscle, anti-LRP4, anti-RyR, anti-AchR) had a sensitivity of 80% (95% CI 0.52-0.96) and a specificity of 88% (95% CI 0.76-0.95) for the diagnosis of myositis, myocarditis, or myasthenia gravis. Conclusion Neuromuscular autoantibodies may serve as a feasible marker to diagnose and potentially predict life-threatening ICI-induced neuromuscular disease. However, brain-reactive autoantibodies are common in both ICI-treated patients with and without irAE-n, hence, their pathogenic significance remains unclear.
Collapse
Affiliation(s)
- Leonie Müller-Jensen
- Klinik und Hochschulambulanz für Neurologie, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt - Universität zu Berlin, Berlin, Germany,Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Samuel Knauss
- Klinik und Hochschulambulanz für Neurologie, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt - Universität zu Berlin, Berlin, Germany,Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Lorena Ginesta Roque
- Klinik und Hochschulambulanz für Neurologie, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt - Universität zu Berlin, Berlin, Germany
| | - Christian Schinke
- Klinik und Hochschulambulanz für Neurologie, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt - Universität zu Berlin, Berlin, Germany,Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Smilla K. Maierhof
- Klinik und Hochschulambulanz für Neurologie, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt - Universität zu Berlin, Berlin, Germany,Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Frederik Bartels
- Klinik und Hochschulambulanz für Neurologie, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt - Universität zu Berlin, Berlin, Germany,Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Carsten Finke
- Klinik und Hochschulambulanz für Neurologie, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt - Universität zu Berlin, Berlin, Germany,Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany,Berlin School of Mind and Brain, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Kristin Rentzsch
- Clinical Immunological Laboratory Prof. Dr. med. Winfried Stöcker, Groß Grönau, Germany
| | - Claas Ulrich
- Hauttumorcentrum, Klinik für Dermatologie, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Raphael Mohr
- Medizinische Klinik mit Schwerpunkt Gastroenterologie und Hepatologie, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Werner Stenzel
- Institut für Neuropathologie, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Matthias Endres
- Klinik und Hochschulambulanz für Neurologie, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt - Universität zu Berlin, Berlin, Germany,NeuroCure Cluster of Excellence, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany,Center for Stroke Research, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany,German Center for Neurodegenerative Diseases (DZNE), partner site, Berlin, Germany,German Center for Cardiovascular Research (DZHK), partner site, Berlin, Germany
| | - Wolfgang Boehmerle
- Klinik und Hochschulambulanz für Neurologie, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt - Universität zu Berlin, Berlin, Germany,Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany,NeuroCure Cluster of Excellence, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany,*Correspondence: Wolfgang Boehmerle,
| | - Petra Huehnchen
- Klinik und Hochschulambulanz für Neurologie, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt - Universität zu Berlin, Berlin, Germany,Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany,NeuroCure Cluster of Excellence, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
236
|
Radiotherapy, PARP Inhibition, and Immune-Checkpoint Blockade: A Triad to Overcome the Double-Edged Effects of Each Single Player. Cancers (Basel) 2023; 15:cancers15041093. [PMID: 36831435 PMCID: PMC9954050 DOI: 10.3390/cancers15041093] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 02/05/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023] Open
Abstract
Radiotherapy and, more recently, PARP inhibitors (PARPis) and immune-checkpoint inhibitors represent effective tools in cancer therapy. Radiotherapy exerts its effects not only by damaging DNA and inducing tumor cell death, but also stimulating anti-tumor immune responses. PARPis are known to exert their therapeutic effects by inhibiting DNA repair, and they may be used in combination with radiotherapy. Both radiotherapy and PARPis modulate inflammatory signals and stimulate type I IFN (IFN-I)-dependent immune activation. However, they can also support the development of an immunosuppressive tumor environment and upregulate PD-L1 expression on tumor cells. When provided as monotherapy, immune-checkpoint inhibitors (mainly antibodies to CTLA-4 and the PD-1/PD-L1 axis) result particularly effective only in immunogenic tumors. Combinations of immunotherapy with therapies that favor priming of the immune response to tumor-associated antigens are, therefore, suitable strategies. The widely explored association of radiotherapy and immunotherapy has confirmed this benefit for several cancers. Association with PARPis has also been investigated in clinical trials. Immunotherapy counteracts the immunosuppressive effects of radiotherapy and/or PARPis and synergies with their immunological effects, promoting and unleashing immune responses toward primary and metastatic lesions (abscopal effect). Here, we discuss the beneficial and counterproductive effects of each therapy and how they can synergize to overcome single-therapy limitations.
Collapse
|
237
|
Lin L, Yang F, Lin G, Chen X. Thyroid Dysfunction in Non-Small Cell Lung Cancer With Immune Checkpoint Inhibitors: A Meta-Analysis. J Clin Pharmacol 2023; 63:210-218. [PMID: 36083133 DOI: 10.1002/jcph.2150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 08/29/2022] [Indexed: 01/18/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have been established as the cornerstone for advanced non-small cell lung cancer, while thyroid adverse events (AEs) associated with ICIs have not been systematically documented. Therefore, we performed a meta-analysis to evaluate the effect of ICI applications on the thyroid of patients with non-small cell lung cancer. We performed a systematic search of PubMed, the Cochrane Library, Web of Science, and Embase for eligible randomized controlled trials up to December 2021. Clinical trials reporting thyroid AEs including hypothyroidism, hyperthyroidism, and thyroiditis were enrolled. The I2 statistic was also calculated to quantify the heterogeneity. Data were evaluated as risk ratio (RR) and corresponding 95%CIs. A total of 10 randomized clinical trials involving 6154 patients were included in this meta-analysis. ICI application was found to have a statistically significant higher risk of all grade hypothyroidism (RR, 7.03; P < 0.0001), hyperthyroidism (RR, 4.88; P < 0.0001), and thyroiditis (RR, 6.58; P = 0.0014) compared with the chemotherapy group. Moreover, we demonstrated that second-line therapy significantly increased the risk of all-grade hypothyroidism (RR, 7.03 [95%CI, 4.69-10.55]) and hyperthyroidism (RR, 4.88 [95%CI, 3.11-7.65]). Our meta-analysis manifested that regimens with ICIs may significantly increase all grades of hypothyroidism, hyperthyroidism, and thyroiditis. ICIs may induce the occurrence and exacerbation of endocrine AEs compared with chemotherapy.
Collapse
Affiliation(s)
- Lanlan Lin
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, People's Republic of China
| | - Fan Yang
- Department of Respiratory Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian, People's Republic of China
| | - Guofu Lin
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, People's Republic of China
| | - Xiangqi Chen
- Department of Respiratory Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian, People's Republic of China
| |
Collapse
|
238
|
Liu TW, Gammon ST, Yang P, Ma W, Wang J, Piwnica-Worms D. Inhibition of myeloperoxidase enhances immune checkpoint therapy for melanoma. J Immunother Cancer 2023; 11:e005837. [PMID: 36805920 PMCID: PMC9944647 DOI: 10.1136/jitc-2022-005837] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2023] [Indexed: 02/19/2023] Open
Abstract
BACKGROUND The presence of a highly immunosuppressive tumor microenvironment has limited the success of immune checkpoint therapy (ICT). Immune suppressing myeloid cells with increased production of reactive oxygen species are critical drivers of this immunosuppressive tumor microenvironment. Strategies to limit these immune suppressing myeloid cells are needed to enhance response to ICT. METHODS To evaluate the contribution of myeloperoxidase (MPO), a myeloid lineage-restricted enzyme and a major source of reactive oxygen species, to mediating ICT response, we compared treatment outcome and immune composition in wild-type, MPO-deficient (MPO -/- ), and MPO inhibitor-treated wild-type mice using established primary melanoma models. RESULTS Tumor growth and survival studies demonstrated that either host deficiency (MPO -/- ) or pharmacological inhibition of MPO enhanced ICT response in two preclinical models of established primary melanoma in aged animals. The tumor microenvironment and systemic immune landscape underwent striking changes in infiltration of myeloid cells, T cells, B cells, and dendritic cells in MPO -/- mice; furthermore, a significant increase in myeloid cells was observed in ICT non-responders. The contribution of CD4+ T cells and NK cells during ICT response also changed in MPO -/- mice. Interestingly, MPO enzymatic activity, but not protein, was increased in CD11b+Ly6G+ myeloid cells isolated from marrow, spleen, and peritoneal cavities of mice bearing untreated melanoma, indicating systemic activation of innate immunity. Notably, repurposing MPO-specific inhibitors (verdiperstat, AZD5904) in combination with ICT pointedly enhanced response rates above ICT alone. Indeed, long-term survival was 100% in the YUMM3.3 melanoma model on treatment with verdiperstat plus ICT. CONCLUSION MPO contributes to ICT resistance in established melanoma. Repurposing MPO-specific inhibitors may provide a promising therapeutic strategy to enhance ICT response.
Collapse
Affiliation(s)
- Tracy W Liu
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
| | - Seth T Gammon
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ping Yang
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Wencai Ma
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jing Wang
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - David Piwnica-Worms
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
239
|
Abiko K, Hamanishi J, Matsumura N, Mandai M. Dynamic host immunity and PD-L1/PD-1 blockade efficacy: developments after "IFN-γ from lymphocytes induces PD-L1 expression and promotes progression of ovarian cancer". Br J Cancer 2023; 128:461-467. [PMID: 36068276 PMCID: PMC9938281 DOI: 10.1038/s41416-022-01960-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/01/2022] [Accepted: 08/11/2022] [Indexed: 11/09/2022] Open
Abstract
In the article titled "IFN-γ from lymphocytes induces PD-L1 expression and promotes progression of ovarian cancer" in 2015, we showed that PD-L1 expression is induced by IFN-γ from lymphocytes in the tumour microenvironment. This article proposed that PD-L1 expression in cancer cells is not stable but varies among cases, or even within a case, which is influenced by the stromal infiltration of cytotoxic lymphocytes. Immune-checkpoint inhibitors, especially anti-PD-1/PD-L1 therapies, are now widely used to treat various types of cancer. Predictive biomarkers for the efficacy of immune-checkpoint inhibitors include PD-L1 expression, MSI/mismatch repair deficiency and high tumour mutation burden. However, clinical trials have proven that their use in ovarian cancer is still challenging. Reliable biomarkers and new treatment strategies may be sought by elucidating the complex immune microenvironment of ovarian cancer. Although the interaction between cytotoxic lymphocytes and PD-1/PD-L1 on tumour cells is at the centre of therapeutic targets, other immune checkpoints and various immunosuppressive cells also play important roles in ovarian cancer. Targeting these role players in combination with PD-1/PD-L1 blockade may be a promising therapeutic strategy.
Collapse
Affiliation(s)
- Kaoru Abiko
- Department of Obstetrics and Gynecology, National Hospital Organization Kyoto Medical Center, Kyoto, Japan.
| | - Junzo Hamanishi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Noriomi Matsumura
- Department of Obstetrics and Gynecology, Kindai University Faculty of Medicine, Osaka-sayama, Osaka prefecture, Japan
| | - Masaki Mandai
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
240
|
Tan P, Chen X, Zhang H, Wei Q, Luo K. Artificial intelligence aids in development of nanomedicines for cancer management. Semin Cancer Biol 2023; 89:61-75. [PMID: 36682438 DOI: 10.1016/j.semcancer.2023.01.005] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/28/2022] [Accepted: 01/18/2023] [Indexed: 01/21/2023]
Abstract
Over the last decade, the nanomedicine has experienced unprecedented development in diagnosis and management of diseases. A number of nanomedicines have been approved in clinical use, which has demonstrated the potential value of clinical transition of nanotechnology-modified medicines from bench to bedside. The application of artificial intelligence (AI) in development of nanotechnology-based products could transform the healthcare sector by realizing acquisition and analysis of large datasets, and tailoring precision nanomedicines for cancer management. AI-enabled nanotechnology could improve the accuracy of molecular profiling and early diagnosis of patients, and optimize the design pipeline of nanomedicines by tuning the properties of nanomedicines, achieving effective drug synergy, and decreasing the nanotoxicity, thereby, enhancing the targetability, personalized dosing and treatment potency of nanomedicines. Herein, the advances in AI-enabled nanomedicines in cancer management are elaborated and their application in diagnosis, monitoring and therapy as well in precision medicine development is discussed.
Collapse
Affiliation(s)
- Ping Tan
- Department of Urology, and Department of Radiology, Institute of Urology, and Huaxi MR Research Center (HMRRC), Animal Experimental Center, National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaoting Chen
- Department of Urology, and Department of Radiology, Institute of Urology, and Huaxi MR Research Center (HMRRC), Animal Experimental Center, National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hu Zhang
- Amgen Bioprocessing Centre, Keck Graduate Institute, Claremont, CA 91711, USA
| | - Qiang Wei
- Department of Urology, and Department of Radiology, Institute of Urology, and Huaxi MR Research Center (HMRRC), Animal Experimental Center, National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Kui Luo
- Department of Urology, and Department of Radiology, Institute of Urology, and Huaxi MR Research Center (HMRRC), Animal Experimental Center, National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
241
|
Advances in pharmacokinetics and pharmacodynamics of PD-1/PD-L1 inhibitors. Int Immunopharmacol 2023; 115:109638. [PMID: 36587500 DOI: 10.1016/j.intimp.2022.109638] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/09/2022] [Accepted: 12/22/2022] [Indexed: 12/31/2022]
Abstract
Immune checkpoint inhibitors (ICIs) are a group of drugs designed to improve the therapeutic effects on various types of malignant tumors. Irrespective of monotherapy or combinational therapies as first-line and later-line therapy, ICIs have achieved benefits for various tumors. Programmed cell death protein-1 (PD-1) / ligand 1 (PD-L1) is an immune checkpoint that suppresses antitumor immunity, especially in the tumor microenvironment (TME). PD-1/PD-L1 immune checkpoint inhibitors block tumor-related downregulation of the immune system, thereby enhancing antitumor immunity. In comparison with traditional small-molecule drugs, ICIs exhibit pharmacokinetic characteristics owing to their high molecular weight. Furthermore, different types of ICIs exhibit different pharmacodynamic characteristics. Hence, ICIs have been approved for different indications by the Food and Drug Administration (FDA) and National Medical Products Administration (NMPA). This review summarizes pharmacokinetic and pharmacodynamic studies of PD-1/ PD-L1 inhibitors to provide a reference for rational clinical application.
Collapse
|
242
|
Injectable Polypeptide Hydrogel Depots Containing Dual Immune Checkpoint Inhibitors and Doxorubicin for Improved Tumor Immunotherapy and Post-Surgical Tumor Treatment. Pharmaceutics 2023; 15:pharmaceutics15020428. [PMID: 36839750 PMCID: PMC9965187 DOI: 10.3390/pharmaceutics15020428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/14/2023] [Accepted: 01/19/2023] [Indexed: 01/31/2023] Open
Abstract
In this work, we developed a strategy for local chemo-immunotherapy through simultaneous incorporation of dual immune checkpoint blockade (ICB) antibodies, anti-cytotoxic T-lymphocyte-associated protein 4 (aCTLA-4) and anti-programmed cell death protein 1 (aPD-1), and a chemotherapy drug, doxorubicin (Dox), into a thermo-gelling polypeptide hydrogel. The hydrogel encapsulating Dox or IgG model antibody showed sustained release profiles for more than 12 days in vitro, and the drug release and hydrogel degradation were accelerated in the presence of enzymes. In comparison to free drug solutions or hydrogels containing Dox or antibodies only, the Dox/aCTLA-4/aPD-1 co-loaded hydrogel achieved improved tumor suppression efficiency, strengthened antitumor immune response, and prolonged animal survival time after peritumoral injection into mice bearing B16F10 melanoma. Additionally, after injection of Dox/aCTLA-4/aPD-1 co-loaded hydrogel into the surgical site following tumor resection, a significantly enhanced inhibition on tumor reoccurrence was demonstrated. Thus, the polypeptide hydrogel-based chemo-immunotherapy strategy has potential in anti-tumor therapy and the prevention of tumor reoccurrence.
Collapse
|
243
|
Du X, Wen S, Shi R, Xia J, Wang R, Zhang Y, Pan B, Wu X, Zhu W, Feng J, Wang X, Shen B. Peripheral blood lymphocytes differentiation patterns in responses / outcomes to immune checkpoint blockade therapies in non-small cell lung cancer: a retrospective study. BMC Cancer 2023; 23:83. [PMID: 36698098 PMCID: PMC9875514 DOI: 10.1186/s12885-023-10502-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 01/02/2023] [Indexed: 01/26/2023] Open
Abstract
OBJECTIVES Programmed Cell Death-1/ Programmed Death-ligand 1 (PD-1 / PD-L1) inhibitor therapies targeting immunocytes induce persistent tumor remission in various cancers. However, the appropriate biomarkers for the therapeutic efficacy of PD-L1 and PD-1 blockade remain elusive. MATERIALS AND METHODS For a comprehensive analysis of peri-treatment lymphocyte differentiation, in the current study, we enrolled 146 non-small cell lung cancer patients who received α-PD-1 therapies for exploring the peripheral blood lymphocyte differentiation pattern at baseline and post-treatment (dynamic changes) by flow cytometry. RESULTS At baseline, CD4+ / CD8+ T cell ratio predicts good responses and outcomes, but activated T cell and cytotoxic T cell counts predict poor responses and outcomes. And for dynamic changes, after 6 weeks of immune checkpoint blockade (ICB) treatment, compared with baseline level, the elevation of total T and B cell counts indicate poor responses, and total T and TH cell counts indicate poor prognosis while activated T cell predicts good prognosis. And after 12 weeks, elevated total lymphocyte, cytotoxic T cell counts, and decreased total T cell counts and CD4+ / CD8+ T cell ratio predict good responses / outcomes. Our clinical predicting model shows good performance in predicting ICB treatment responses / outcomes. CONCLUSION Patients with favorable clinical responses / outcomes have distinctive peripheral blood immunocyte differentiation characteristics, indicating the potential of utilizing the peripheral immunocyte differentiation patterns for predicting ICB responses / outcomes.
Collapse
Affiliation(s)
- Xiaoyue Du
- grid.452509.f0000 0004 1764 4566Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Shaodi Wen
- grid.452509.f0000 0004 1764 4566Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Run Shi
- grid.412676.00000 0004 1799 0784Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jingwei Xia
- grid.452509.f0000 0004 1764 4566Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Ruotong Wang
- grid.452509.f0000 0004 1764 4566Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Yihan Zhang
- grid.452509.f0000 0004 1764 4566Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Banzhou Pan
- grid.452509.f0000 0004 1764 4566Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Xiaoliu Wu
- grid.452509.f0000 0004 1764 4566Flow Cytometry Core Facility, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Wei Zhu
- grid.440785.a0000 0001 0743 511XSchool of Medicine, Jiangsu University, Zhenjiang, China
| | - Jifeng Feng
- grid.452509.f0000 0004 1764 4566Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Xin Wang
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China.
| | - Bo Shen
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China.
| |
Collapse
|
244
|
Nicolaides S, Boussioutas A. Immune-Related Adverse Events of the Gastrointestinal System. Cancers (Basel) 2023; 15:cancers15030691. [PMID: 36765649 PMCID: PMC9913287 DOI: 10.3390/cancers15030691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/19/2023] [Accepted: 01/21/2023] [Indexed: 01/24/2023] Open
Abstract
Immune checkpoint inhibitors (ICI) are a form of immunotherapy that have revolutionized the treatment of a number of cancers. Specifically, they are antibodies targeted against established and emerging immune checkpoints, such as cytotoxic T-cell antigen 4 (CTLA4), programmed cell death ligand 1 (PD-L1) and programmed cell death 1 protein (PD-1) on CD8-positive T cells, which promote the destruction of tumor cells. While the immune checkpoint inhibitors are very effective in the treatment of a number of cancers, their use is limited by serious and in some cases life-threatening immune-related adverse events. While these involve many organs, one of the most prevalent serious adverse events is immune checkpoint inhibitor colitis, occurring in a significant proportion of patients treated with this therapy. In this review, we aim to broadly describe the immune-related adverse events known to occur within the gastrointestinal system and the potential role played by the intestinal microbiome.
Collapse
Affiliation(s)
- Steven Nicolaides
- Department of Gastroenterology, Western Health, Melbourne, VIC 3011, Australia
- Department of Gastroenterology, The Alfred, Melbourne, VIC 3004, Australia
| | - Alex Boussioutas
- Department of Gastroenterology, The Alfred, Melbourne, VIC 3004, Australia
- Department of Medicine, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Melbourne, VIC 3050, Australia
- Correspondence:
| |
Collapse
|
245
|
Iaccarino A, Nacchio M, Acanfora G, Pisapia P, Malapelle U, Bellevicine C, Troncone G, Vigliar E. Multiple predictive biomarker testing in melanoma: Another challenge in identifying the optimal approach on cytological samples. Cytopathology 2023; 34:198-203. [PMID: 36658094 DOI: 10.1111/cyt.13211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 01/12/2023] [Indexed: 01/21/2023]
Abstract
BACKGROUND The management of cutaneous melanoma has changed dramatically in recent years thanks to the development of tyrosine kinase and immune-checkpoint inhibitors (ICIs). Thus, multiple biomarker testing is becoming ever more important for the identification of patients who are potentially eligible for these treatments. One reliable approach to the molecular evaluation of metastatic melanoma is fine needle cytology (FNC). To examine the utility of this approach for assessing PD-L1 expression levels, we evaluated the cellular adequacy of residual cell block (CB) material from metastatic melanomas that were previously tested for BRAF and NRAS mutations. METHODS We retrieved from our internal archives a series of FNC samples of metastatic melanoma that had been subjected to molecular testing on residual CB material or a dedicated needle rinse between January 2016 and July 2022. Real-time polymerase chain reaction was used to assess BRAF and NRAS status, and an SP263 assay was employed to ascertain PD-L1 expression levels. RESULTS Overall, n = 19 cases were selected. Of these, 11 (57.9%) cases revealed a BRAF exon 15 p.V600E mutation, one case (5.3%) revealed NRAS mutation, and seven cases (36.8%) showed no mutations. Regarding PD-L1 assessment, 16/19 (84.2%) cases were deemed adequate, meaning they contained at least 100 viable cells. CONCLUSIONS We highlighted the feasibility of assessing PD-L1 expression levels in residual CB material from metastatic melanomas previously tested for BRAF and NRAS mutations. Moreover, we pointed out that FNC needle rinses may be an alternative source of nucleic acids for molecular testing, preserving CB material for immunocytochemistry evaluation.
Collapse
Affiliation(s)
- Antonino Iaccarino
- Department of Public Health, University of Naples "Federico II", Naples, Italy
| | - Mariantonia Nacchio
- Department of Public Health, University of Naples "Federico II", Naples, Italy
| | - Gennaro Acanfora
- Department of Public Health, University of Naples "Federico II", Naples, Italy
| | - Pasquale Pisapia
- Department of Public Health, University of Naples "Federico II", Naples, Italy
| | - Umberto Malapelle
- Department of Public Health, University of Naples "Federico II", Naples, Italy
| | - Claudio Bellevicine
- Department of Public Health, University of Naples "Federico II", Naples, Italy
| | - Giancarlo Troncone
- Department of Public Health, University of Naples "Federico II", Naples, Italy
| | - Elena Vigliar
- Department of Public Health, University of Naples "Federico II", Naples, Italy
| |
Collapse
|
246
|
Kaneko S, Asahina Y, Nakagawa M, Murakawa M, Miyazaki Y, Asakage T, Fukuda S, Namiki T, Kano Y, Nagata M, Tsuchiya J, Miyoshi M, Kitahata-Kawai F, Nitta S, Itsui Y, Kakinuma S, Okamoto R. Factors associated with liver injury and prognosis in advanced cancer patients treated with immune checkpoint inhibitors. Hepatol Res 2023; 53:450-459. [PMID: 36626292 DOI: 10.1111/hepr.13878] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/21/2022] [Accepted: 01/05/2023] [Indexed: 01/11/2023]
Abstract
AIM The use of immune checkpoint inhibitors (ICIs) has increased remarkably, and immune-related adverse events (irAEs) have also increased. This study aimed to identify factors associated with immune-related liver injury (irLI), and the relationship between the grades of irLI and overall survival (OS) in patients treated with ICIs. METHODS A total of 571 patients who had been treated for advanced malignancies with ICIs between January 2015 and March 2022 were retrospectively recruited. The presence of liver injury was determined by the aspartate aminotransferase and alanine aminotransferase elevation. The irLI grading was based on Common Terminology Criteria for Adverse Events version 5.0. RESULTS A total of 50 (8.8%) patients had grade ≥2 irLI and 24 (4.2%) had grade ≥3 irLI. Treatment with anti-cytotoxic T-lymphocyte-associated protein-4 agents and baseline grade 1 aspartate aminotransferase/alanine aminotransferase elevation were independent predictive factors of grade ≥2 irLI. Treatment with anti-cytotoxic T-lymphocyte-associated protein-4 was the only independent predictive factor of grade ≥3 irLI. The median OS for patients who experienced any irAEs was significantly longer than of those without irAEs (hazard ratio 0.503, 95% CI 0.398-0.636, p < 0.001). The median OS in patients with grade ≥2 irLI was significantly longer (HR 0.570, 95% CI 0.387-0.838, p = 0.022). There was no significant difference between the median OS in patients with grade ≥3 irLI and the others (p = 0.11). CONCLUSION The incidence of irLI was significantly higher in patients treated with anti-cytotoxic T-lymphocyte-associated protein-4 agents. Even in patients with pre-existing grade 1 aspartate aminotransferase/alanine aminotransferase elevation, appropriate follow-up and control of the irLI can improve the prognosis.
Collapse
Affiliation(s)
- Shun Kaneko
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yasuhiro Asahina
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan.,Department of Liver Disease Control, Tokyo Medical and Dental University, Tokyo, Japan
| | - Mina Nakagawa
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Miyako Murakawa
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yasunari Miyazaki
- Department of Respiratory Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takahiro Asakage
- Department of Head and Neck Surgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shohei Fukuda
- Department of Urology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takeshi Namiki
- Department of Dermatology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yoshihito Kano
- Department of Clinical Oncology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masashi Nagata
- Department of Pharmacy, Tokyo Medical and Dental University, Tokyo, Japan
| | - Jun Tsuchiya
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masato Miyoshi
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Fukiko Kitahata-Kawai
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Sayuri Nitta
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yasuhiro Itsui
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Sei Kakinuma
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ryuichi Okamoto
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
247
|
Burkard M, Niessner H, Leischner C, Piotrowsky A, Renner O, Marongiu L, Lauer UM, Busch C, Sinnberg T, Venturelli S. High-Dose Ascorbate in Combination with Anti-PD1 Checkpoint Inhibition as Treatment Option for Malignant Melanoma. Cells 2023; 12:254. [PMID: 36672190 PMCID: PMC9857291 DOI: 10.3390/cells12020254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/14/2022] [Accepted: 01/03/2023] [Indexed: 01/11/2023] Open
Abstract
Ascorbate acts as a prooxidant when administered parenterally at high supraphysiological doses, which results in the generation of hydrogen peroxide in dependence on oxygen. Most cancer cells are susceptible to the emerging reactive oxygen species (ROS). Accordingly, we evaluated high-dose ascorbate for the treatment of the B16F10 melanoma model. To investigate the effects of ascorbate on the B16F10 cell line in vitro, viability, cellular impedance, and ROS production were analyzed. In vivo, C57BL/6NCrl mice were subcutaneously injected into the right flank with B16F10 cells and tumor-bearing mice were treated intraperitoneally with ascorbate (3 g/kg bodyweight), immunotherapy (anti-programmed cell death protein 1 (PD1) antibody J43; 2 mg/kg bodyweight), or both treatments combined. The efficacy and toxicity were analyzed by measuring the respective tumor sizes and mouse weights accompanied by histological analysis of the protein levels of proliferating cell nuclear antigen (Pcna), glucose transporter 1 (Glut-1), and CD3. Treatment of B16F10 melanoma-carrying mice with high-dose ascorbate yielded plasma levels in the pharmacologically effective range, and ascorbate showed efficacy as a monotherapy and when combined with PD1 inhibition. Our data suggest the applicability of ascorbate as an additional therapeutic agent that can be safely combined with immunotherapy and has the potential to potentiate anti-PD1-based immune checkpoint blockades.
Collapse
Affiliation(s)
- Markus Burkard
- Department of Nutritional Biochemistry, Institute of Nutritional Sciences, University of Hohenheim, Garbenstraße 30, 70599 Stuttgart, Germany
| | - Heike Niessner
- Department of Nutritional Biochemistry, Institute of Nutritional Sciences, University of Hohenheim, Garbenstraße 30, 70599 Stuttgart, Germany
- Division of Dermatooncology, Department of Dermatology, University of Tuebingen, Liebermeisterstraße 25, 72076 Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies”, 72076 Tuebingen, Germany
| | - Christian Leischner
- Department of Nutritional Biochemistry, Institute of Nutritional Sciences, University of Hohenheim, Garbenstraße 30, 70599 Stuttgart, Germany
| | - Alban Piotrowsky
- Department of Nutritional Biochemistry, Institute of Nutritional Sciences, University of Hohenheim, Garbenstraße 30, 70599 Stuttgart, Germany
| | - Olga Renner
- Department of Nutritional Biochemistry, Institute of Nutritional Sciences, University of Hohenheim, Garbenstraße 30, 70599 Stuttgart, Germany
| | - Luigi Marongiu
- Department of Nutritional Biochemistry, Institute of Nutritional Sciences, University of Hohenheim, Garbenstraße 30, 70599 Stuttgart, Germany
- Department of Internal Medicine VIII, University Hospital Tuebingen, Otfried-Mueller-Straße 10, 72076 Tuebingen, Germany
| | - Ulrich M. Lauer
- Department of Internal Medicine VIII, University Hospital Tuebingen, Otfried-Mueller-Straße 10, 72076 Tuebingen, Germany
| | - Christian Busch
- Dermatologie zum Delfin, Stadthausstraße 12, 8400 Winterthur, Switzerland
| | - Tobias Sinnberg
- Division of Dermatooncology, Department of Dermatology, University of Tuebingen, Liebermeisterstraße 25, 72076 Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies”, 72076 Tuebingen, Germany
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Sascha Venturelli
- Department of Nutritional Biochemistry, Institute of Nutritional Sciences, University of Hohenheim, Garbenstraße 30, 70599 Stuttgart, Germany
- Department of Vegetative and Clinical Physiology, Institute of Physiology, University of Tuebingen, Wilhelmstraße 56, 72074 Tuebingen, Germany
| |
Collapse
|
248
|
Wang Y, Yan A, Song D, Dong C, Rao M, Gao Y, Qi R, Ma X, Wang Q, Xu H, Liu H, Han J, Duan M, Liu S, Yu X, Zong M, Feng J, Jiao J, Zhang H, Li M, Yu B, Wang Y, Meng F, Ni X, Li Y, Shen Z, Sun B, Shao X, Zhao H, Zhao Y, Li R, Zhang Y, Du G, Lu J, You C, Jiang H, Zhang L, Wang L, Dou C, Liu Z, Zhao J. Biparatopic antibody BA7208/7125 effectively neutralizes SARS-CoV-2 variants including Omicron BA.1-BA.5. Cell Discov 2023; 9:3. [PMID: 36609558 PMCID: PMC9822811 DOI: 10.1038/s41421-022-00509-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 12/07/2022] [Indexed: 01/09/2023] Open
Abstract
SARS-CoV-2 Omicron subvariants have demonstrated extensive evasion from monoclonal antibodies (mAbs) developed for clinical use, which raises an urgent need to develop new broad-spectrum mAbs. Here, we report the isolation and analysis of two anti-RBD neutralizing antibodies BA7208 and BA7125 from mice engineered to produce human antibodies. While BA7125 showed broadly neutralizing activity against all variants except the Omicron sublineages, BA7208 was potently neutralizing against all tested SARS-CoV-2 variants (including Omicron BA.1-BA.5) except Mu. By combining BA7208 and BA7125 through the knobs-into-holes technology, we generated a biparatopic antibody BA7208/7125 that was able to neutralize all tested circulating SARS-CoV-2 variants. Cryo-electron microscopy structure of these broad-spectrum antibodies in complex with trimeric Delta and Omicron spike indicated that the contact residues are highly conserved and had minimal interactions with mutational residues in RBD of current variants. In addition, we showed that administration of BA7208/7125 via the intraperitoneal, intranasal, or aerosol inhalation route showed potent therapeutic efficacy against Omicron BA.1 and BA.2 in hACE2-transgenic and wild-type mice and, separately, effective prophylaxis. BA7208/7125 thus has the potential to be an effective candidate as an intervention against COVID-19.
Collapse
Affiliation(s)
- Yanqun Wang
- grid.470124.4State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong China ,grid.410737.60000 0000 8653 1072GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong China
| | - An Yan
- grid.263817.90000 0004 1773 1790Cryo-electron Microscopy Center, Southern University of Science and Technology, Shenzhen, Guangdong China
| | - Deyong Song
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, Shandong, China
| | - Chuangchuang Dong
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, Shandong, China
| | - Muding Rao
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, Shandong, China
| | - Yuanzhu Gao
- grid.263817.90000 0004 1773 1790Cryo-electron Microscopy Center, Southern University of Science and Technology, Shenzhen, Guangdong China
| | - Ruxi Qi
- grid.263817.90000 0004 1773 1790Cryo-electron Microscopy Center, Southern University of Science and Technology, Shenzhen, Guangdong China
| | - Xiaomin Ma
- grid.263817.90000 0004 1773 1790Cryo-electron Microscopy Center, Southern University of Science and Technology, Shenzhen, Guangdong China
| | - Qiaoping Wang
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, Shandong, China
| | - Hongguang Xu
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, Shandong, China
| | - Hong Liu
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, Shandong, China
| | - Jing Han
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, Shandong, China
| | - Maoqin Duan
- grid.410749.f0000 0004 0577 6238Division of Monoclonal Antibodies, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Shuo Liu
- grid.410749.f0000 0004 0577 6238Division of Monoclonal Antibodies, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Xiaoping Yu
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, Shandong, China
| | - Mengqi Zong
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, Shandong, China
| | - Jianxia Feng
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, Shandong, China
| | - Jie Jiao
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, Shandong, China
| | - Huimin Zhang
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, Shandong, China
| | - Min Li
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, Shandong, China
| | - Beibei Yu
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, Shandong, China
| | | | | | | | - Ying Li
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, Shandong, China
| | - Zhenduo Shen
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, Shandong, China
| | - Baiping Sun
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, Shandong, China
| | - Xin Shao
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, Shandong, China
| | - Haifeng Zhao
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, Shandong, China
| | - Yanyan Zhao
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, Shandong, China
| | - Rui Li
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, Shandong, China
| | - Yanan Zhang
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, Shandong, China
| | - Guangying Du
- State Key Laboratory of Long-acting and Targeting Drug Delivery System, Shandong Luye Pharmaceutical Co. Ltd, Yantai, Shandong, China
| | - Jun Lu
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, Shandong, China
| | - Chunna You
- State Key Laboratory of Long-acting and Targeting Drug Delivery System, Shandong Luye Pharmaceutical Co. Ltd, Yantai, Shandong, China
| | - Hua Jiang
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, Shandong, China
| | - Lu Zhang
- Health and Quarantine Laboratory, Guangzhou Customs District Technology Centre, Guangzhou, Guangdong, China
| | - Lan Wang
- grid.410749.f0000 0004 0577 6238Division of Monoclonal Antibodies, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Changlin Dou
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, Shandong, China
| | - Zheng Liu
- grid.263817.90000 0004 1773 1790Cryo-electron Microscopy Center, Southern University of Science and Technology, Shenzhen, Guangdong China
| | - Jincun Zhao
- grid.470124.4State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong China ,Guangzhou Laboratory, Bio-Island, Guangzhou, Guangdong, China ,grid.413419.a0000 0004 1757 6778Institute of Infectious disease, Guangzhou Eighth People’s Hospital of Guangzhou Medical University, Guangzhou, Guangdong China ,grid.440637.20000 0004 4657 8879Shanghai Institute for Advanced Immunochemical Studies, School of Life Science and Technology, ShanghaiTech University, Shanghai, China ,grid.263817.90000 0004 1773 1790Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital; The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong China
| |
Collapse
|
249
|
Blake MK, O’Connell P, Aldhamen YA. Fundamentals to therapeutics: Epigenetic modulation of CD8 + T Cell exhaustion in the tumor microenvironment. Front Cell Dev Biol 2023; 10:1082195. [PMID: 36684449 PMCID: PMC9846628 DOI: 10.3389/fcell.2022.1082195] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 12/16/2022] [Indexed: 01/06/2023] Open
Abstract
In the setting of chronic antigen exposure in the tumor microenvironment (TME), cytotoxic CD8+ T cells (CTLs) lose their immune surveillance capabilities and ability to clear tumor cells as a result of their differentiation into terminally exhausted CD8+ T cells. Immune checkpoint blockade (ICB) therapies reinvigorate exhausted CD8+ T cells by targeting specific inhibitory receptors, thus promoting their cytolytic activity towards tumor cells. Despite exciting results with ICB therapies, many patients with solid tumors still fail to respond to such therapies and patients who initially respond can develop resistance. Recently, through new sequencing technologies such as the assay for transposase-accessible chromatin with sequencing (ATAC-seq), epigenetics has been appreciated as a contributing factor that enforces T cell differentiation toward exhaustion in the TME. Importantly, specific epigenetic alterations and epigenetic factors have been found to control CD8+ T cell exhaustion phenotypes. In this review, we will explain the background of T cell differentiation and various exhaustion states and discuss how epigenetics play an important role in these processes. Then we will outline specific epigenetic changes and certain epigenetic and transcription factors that are known to contribute to CD8+ T cell exhaustion. We will also discuss the most recent methodologies that are used to study and discover such epigenetic modulations. Finally, we will explain how epigenetic reprogramming is a promising approach that might facilitate the development of novel exhausted T cell-targeting immunotherapies.
Collapse
Affiliation(s)
| | | | - Yasser A. Aldhamen
- Department of Microbiology and Molecular Genetics, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
250
|
Guan J, Liu H, Chai Y, Yu J, Yao J, Wang J, Pan Z, Zhang J, Zhou Y, Liu H, Yao S, Qi J, Feng H, Gao GF, Wang Q, Shi Y, Tan S. Characterization of the high-affinity anti-CTLA-4 monoclonal antibody JS007 for immune checkpoint therapy of cancer. MAbs 2023; 15:2153409. [PMID: 36511654 PMCID: PMC9754112 DOI: 10.1080/19420862.2022.2153409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) is a critical inhibitory checkpoint molecule, and monoclonal antibodies (mAbs) targeting CTLA-4 that restore anti-tumor T cell immunity have achieved clinical success. Here, we report a humanized IgG1 mAb, namely JS007, with high binding affinity to CTLA-4. JS007 shows superior binding affinity and T-cell activating efficiency over ipilimumab. Moreover, it demonstrates substantial in vivo tumor suppression efficacy at low doses. The crystal structure of JS007/CTLA-4 complex (PDB: 8HIT) shows JS007 adopts a heavy-chain-dominant binding mode, and mainly contacts the BC loop, DE loop and FG loop of CTLA-4. Notably, two Tyr residues (VH-Y100 and VL-Y32) from the complementarity-determining region loops insert into the two cavities formed by the residues from the loops of CTLA-4, which may contribute to the stabilization of the binding. Comparative analysis with other anti-CTLA-4 mAbs indicates that the double "wedge-into-hole" binding mode is unique for JS007 and may be responsible for the high-affinity binding to CTLA-4. These findings have provided an important molecular understanding of the high-affinity CTLA-4 blockade mAbs and shed light on future development of agents targeting CTLA-4.
Collapse
Affiliation(s)
- Jiawei Guan
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), China,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Hongchuan Liu
- Department of Antibody Discovery and Engineering, Shanghai Junshi Biosciences Co Ltd, Shanghai, China
| | - Yan Chai
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), China
| | - Jie Yu
- Pilot National Laboratory for Marine Science and Technology (Qingdao), Shandong, China
| | - Jian Yao
- Department of Antibody Discovery and Engineering, Shanghai Junshi Biosciences Co Ltd, Shanghai, China
| | - Jing Wang
- Department of Antibody Discovery and Engineering, Shanghai Junshi Biosciences Co Ltd, Shanghai, China
| | - Zhiwei Pan
- Department of Antibody Discovery and Engineering, Shanghai Junshi Biosciences Co Ltd, Shanghai, China
| | - Jing Zhang
- Department of Antibody Discovery and Engineering, Shanghai Junshi Biosciences Co Ltd, Shanghai, China
| | - Yuehua Zhou
- Department of Antibody Discovery and Engineering, Shanghai Junshi Biosciences Co Ltd, Shanghai, China
| | - Hui Liu
- Department of Antibody Discovery and Engineering, Shanghai Junshi Biosciences Co Ltd, Shanghai, China
| | - Sheng Yao
- Department of Antibody Discovery and Engineering, Shanghai Junshi Biosciences Co Ltd, Shanghai, China
| | - Jianxun Qi
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), China
| | - Hui Feng
- Department of Antibody Discovery and Engineering, Shanghai Junshi Biosciences Co Ltd, Shanghai, China
| | - George F. Gao
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), China,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Qihui Wang
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), China,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China,CONTACT Qihui Wang
| | - Yi Shi
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), China,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Shuguang Tan
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), China,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|