101
|
Kuronishi M, Ozawa Y, Kimura T, Li SD, Kato Y. Development of a Microvessel Density Gene Signature and Its Application in Precision Medicine. CANCER RESEARCH COMMUNICATIONS 2025; 5:398-408. [PMID: 39835481 PMCID: PMC11880750 DOI: 10.1158/2767-9764.crc-24-0403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 11/08/2024] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
SIGNIFICANCE A novel gene signature for MVD was developed. This MVD gene score enables the estimation of MVD, reflecting the sensitivity to antiangiogenic inhibitors, in transcriptomic datasets. We demonstrated the utility of the MVD gene score together with a T cell-inflamed gene signature for potential future use as a clinical biomarker.
Collapse
Affiliation(s)
| | - Yoichi Ozawa
- Tsukuba Research Laboratories, Eisai Co., Ltd., Tsukuba, Japan
| | - Takayuki Kimura
- Tsukuba Research Laboratories, Eisai Co., Ltd., Tsukuba, Japan
| | | | - Yu Kato
- Tsukuba Research Laboratories, Eisai Co., Ltd., Tsukuba, Japan
| |
Collapse
|
102
|
Rosso C, Reed M, Walde N, Voutsadakis IA. Radiation therapy in combination with immune checkpoint inhibitors in metastatic lung cancer: Effect of fractionation. J Investig Med 2025; 73:300-309. [PMID: 39534958 DOI: 10.1177/10815589241270439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Immunotherapy with checkpoint inhibitors has improved the outcomes of patients with metastatic lung cancer in recent years. Despite improved prognosis, not all patients respond to treatment. Therapeutic interventions to build on the success of immune checkpoint inhibitors are needed. A retrospective review of patient records for patients who had received immune checkpoint inhibitors in a single cancer center over 4 years was undertaken. Demographic and disease characteristics of patients with metastatic non-small cell lung cancer were recorded. Data on other treatments including chemotherapy and radiation therapy were extracted, and survival outcomes were calculated. Most (81.8%) of the 77 metastatic lung cancer patients examined had received palliative radiation therapy within 3 months of starting immune checkpoint inhibitors. While the survival outcomes of these patients did not differ from patients who had not received radiotherapy, patients who had undergone hypofractionated radiotherapy (defined as one or more fractions of 700 cGy or higher) displayed a better overall survival (OS) than the rest of the cohort. Palliative radiation therapy administered in proximity with immune checkpoint inhibitors immunotherapy had no effect on the OS of metastatic lung cancer patients. However, patients receiving palliative radiotherapy with fractions above 700 cGy showed better OS. Further studies are needed to optimize a combination strategy.
Collapse
Affiliation(s)
| | - Melissa Reed
- Clinical Trials Unit, Sault Area Hospital, Sault Ste. Marie, Ontario, Canada
- Ottawa University School of Medicine, Ottawa, Ontario, Canada
| | - Natalie Walde
- Clinical Trials Unit, Sault Area Hospital, Sault Ste. Marie, Ontario, Canada
| | - Ioannis A Voutsadakis
- Algoma District Cancer Program, Sault Area Hospital, Sault Ste. Marie, ON, Canada
- Section of Internal Medicine, Division of Clinical Sciences, Northern Ontario School of Medicine, Sudbury, ON, Canada
| |
Collapse
|
103
|
Oh CS, Sher EF, Bieber AK. Melanoma in pregnancy. Semin Perinatol 2025; 49:152040. [PMID: 40089319 DOI: 10.1016/j.semperi.2025.152040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/17/2025]
Abstract
Cutaneous melanoma is a malignant neoplasm of melanocytes that most frequently affects the skin. It is the most common malignancy in women of childbearing age, and accounts for almost one-third of all malignancies diagnosed during gestation. The pathophysiology of melanoma, particularly during pregnancy, is not well understood, but there are several ways in which the physiologic state pregnancy may impact melanoma. Based on the available literature, pregnancy does not seem to worsen maternal outcomes with melanoma, and outside of placental and fetal metastases, melanoma does not seem to cause serious obstetric or fetal complications. Treatment of localized melanoma during pregnancy follows guidelines for the general population, but advanced melanoma in pregnancy poses unique challenges given the lack of unifying research and management recommendations. Herein, we review the current literature, highlighting diagnostic clinical pearls and key multidisciplinary management considerations with regard to melanoma in the child-bearing population.
Collapse
Affiliation(s)
- Christina S Oh
- The Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA
| | - Elizabeth F Sher
- The Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA
| | - Amy K Bieber
- The Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
104
|
Hiraoka A, Tada T, Hirooka M, Kariyama K, Tani J, Atsukawa M, Takaguchi K, Itobayashi E, Fukunishi S, Tsuji K, Ishikawa T, Tajiri K, Ohama H, Toyoda H, Ogawa C, Nishimura T, Hatanaka T, Kakizaki S, Kawata K, Naganuma A, Kosaka H, Matono T, Kuroda H, Yata Y, Nishikawa H, Imai M, Aoki T, Ochi H, Tada F, Nakamura S, Nakamura Y, Nouso K, Morishita A, Itokawa N, Okubo T, Arai T, Tsutsui A, Nagano T, Tanaka K, Tanaka H, Koshiyama Y, Kanayama Y, Noritake H, Enomoto H, Kaibori M, Hiasa Y, Kudo M, Kumada T. Efficacy of durvalumab plus tremelimumab treatment for unresectable hepatocellular carcinoma in immunotherapy era clinical practice. Hepatol Res 2025; 55:444-453. [PMID: 39526824 DOI: 10.1111/hepr.14136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/18/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024]
Abstract
AIM Since the development of tremelimumab plus durvalumab (Dur/Tre) for unresectable hepatocellular carcinoma (uHCC), it has been used as not only an initial but also later line treatment in clinical practice. This study aimed to elucidate clinical prognostic factors for progression-free survival (PFS) in Dur/Tre treatment cases. METHODS Enrolled were 183 uHCC patients treated with Dur/Tre from 2023 to May 2024 (median age, 74 years; male patients, 152; Child-Pugh class A:B, 150:33; Barcelona Clinic Liver Cancer stage B:C, 59:124; initial line use, 64). Clinical factors with prognostic influence on PFS in these patients were retrospectively evaluated. RESULTS The median observation period was 7.2 months (interquartile range, 3.2-10.4). History of atezolizumab plus bevacizumab (Atz/Bev) treatment was the only significant prognostic factor for PFS at introduction of Dur/Tre in multivariate analysis (hazard ratio 2.040, p = 0.028) (median PFS: without vs. with = 5.6 vs. 2.7 months, p < 0.001). Although immune-mediated adverse events (imAE) occurrence was only significant in univariate analysis, when objective response and disease control rates were examined according to imAE positivity (any grade) at the time of analysis, those were noted in 14.4% and 39.2%, respectively, of patients without imAE, while in patients with imAE (any grade), they were noted in 18.2% and 56.1%, respectively (p = 0.523 and p = 0.038, respectively). CONCLUSION History of Atz/Bev treatment may be an independent clinical factor for poor PFS at Dur/Tre introduction.
Collapse
Affiliation(s)
- Atsushi Hiraoka
- Department of Gastroenterology, Ehime Prefectural Central Hospital, Matsuyama, Japan
| | - Toshifumi Tada
- Department of Internal Medicine, Japanese Red Cross Himeji Hospital, Himeji, Japan
| | - Masashi Hirooka
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Toon, Japan
| | - Kazuya Kariyama
- Department of Gastroenterology and Liver Disease Center, Okayama City Hospital, Okayama, Japan
| | - Joji Tani
- Department of Gastroenterology and Neurology, Kagawa University, Takamatsu, Japan
| | - Masanori Atsukawa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Nippon Medical School, Tokyo, Japan
| | - Koichi Takaguchi
- Department of Hepatology, Kagawa Prefectural Central Hospital, Takamatsu, Japan
| | - Ei Itobayashi
- Department of Gastroenterology, Asahi General Hospital, Asahi, Japan
| | - Shinya Fukunishi
- Department of Gastroenterology, Hyogo Medical University, Nishinomiya, Japan
| | - Kunihiko Tsuji
- Center of Gastroenterology, Teine Keijinkai Hospital, Sapporo, Japan
| | - Toru Ishikawa
- Department of Gastroenterology, Saiseikai Niigata Hospital, Niigata, Japan
| | - Kazuto Tajiri
- Department of Gastroenterology, Toyama University Hospital, Toyama, Japan
| | - Hideko Ohama
- Department of Gastroenterology, Takarazuka City Hospital, Takarazuka, Japan
| | - Hidenori Toyoda
- Department of Gastroenterology and Hepatology, Ogaki Municipal Hospital, Ogaki, Japan
| | - Chikara Ogawa
- Department of Gastroenterology, Japanese Red Cross Takamatsu Hospital, Takamatsu, Japan
| | - Takashi Nishimura
- Department of Gastroenterology, Hyogo Medical University, Nishinomiya, Japan
| | - Takeshi Hatanaka
- Department of Gastroenterology, Gunma Saiseikai Maebashi Hospital, Maebashi, Japan
| | - Satoru Kakizaki
- Department of Clinical Research, NHO Takasaki General Medical Center, Takasaki, Japan
| | - Kazuhito Kawata
- Hepatology Division, Department of Internal Medicine II, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Atsushi Naganuma
- Department of Gastroenterology, NHO Takasaki General Medical Center, Takasaki, Japan
| | - Hisashi Kosaka
- Department of Hepatobiliary Surgery, Kansai Medical University, Hirakata, Japan
| | - Tomomitsu Matono
- Department of Hepatology, Harima Himeji General Medical Center, Himeji, Japan
| | - Hidekatsu Kuroda
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Iwate Medical University School of Medicine, Morioka, Japan
| | - Yutaka Yata
- Department of Gastroenterology, Hanwa Memorial Hospital, Osaka, Japan
| | - Hiroki Nishikawa
- Department of Gastroenterology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Michitaka Imai
- Department of Gastroenterology, Saiseikai Niigata Hospital, Niigata, Japan
- Department of Gastroenterology, Niigata Prefectural Cancer Center, Niigata, Japan
| | - Tomoko Aoki
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Hironori Ochi
- Hepato-Biliary Center, Matsuyama Red Cross Hospital, Matsuyama, Japan
| | - Fujimasa Tada
- Department of Gastroenterology, Ehime Prefectural Central Hospital, Matsuyama, Japan
| | - Shinichiro Nakamura
- Department of Internal Medicine, Japanese Red Cross Himeji Hospital, Himeji, Japan
| | - Yoshiko Nakamura
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Toon, Japan
| | - Kazuhiro Nouso
- Department of Gastroenterology and Liver Disease Center, Okayama City Hospital, Okayama, Japan
| | - Asahiro Morishita
- Department of Gastroenterology and Neurology, Kagawa University, Takamatsu, Japan
| | - Norio Itokawa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Nippon Medical School, Tokyo, Japan
| | - Tomomi Okubo
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Nippon Medical School, Tokyo, Japan
| | - Taeang Arai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Nippon Medical School, Tokyo, Japan
| | - Akemi Tsutsui
- Department of Hepatology, Kagawa Prefectural Central Hospital, Takamatsu, Japan
| | - Takuya Nagano
- Department of Hepatology, Kagawa Prefectural Central Hospital, Takamatsu, Japan
| | - Kazunari Tanaka
- Center of Gastroenterology, Teine Keijinkai Hospital, Sapporo, Japan
| | - Hironori Tanaka
- Department of Gastroenterology, Takarazuka City Hospital, Takarazuka, Japan
| | - Yuichi Koshiyama
- Department of Gastroenterology and Hepatology, Ogaki Municipal Hospital, Ogaki, Japan
| | - Yuki Kanayama
- Department of Gastroenterology, Gunma Saiseikai Maebashi Hospital, Maebashi, Japan
| | - Hidenao Noritake
- Hepatology Division, Department of Internal Medicine II, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hirayuki Enomoto
- Department of Gastroenterology, Hyogo Medical University, Nishinomiya, Japan
| | - Masaki Kaibori
- Department of Hepatobiliary Surgery, Kansai Medical University, Hirakata, Japan
| | - Yoichi Hiasa
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Toon, Japan
| | - Masatoshi Kudo
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Takashi Kumada
- Department of Nursing, Gifu Kyoritsu University, Ogaki, Japan
| |
Collapse
|
105
|
Song P, Pan G, Zhang Y, Ni Y, Wang Q, Shi J, Peng Y, Jing R, Luo D. Prospects and Challenges of Immunotherapy for Thyroid Cancer. Endocr Pract 2025; 31:373-379. [PMID: 39631664 DOI: 10.1016/j.eprac.2024.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/21/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Thyroid cancer generally boasts a favorable prognosis; however, advanced and refractory cases exhibit aggressive characteristics and resistance to conventional therapies, necessitating the investigation of innovative treatment modalities. Immunotherapy, which harnesses the body's immune system to target cancer cells, has shown considerable promise for specific thyroid cancer subtypes. OBJECTIVE This review article aims to encapsulate the latest advancements in immunotherapy for thyroid cancer, examining its mechanisms, therapeutic efficacy, ongoing challenges, and the potential benefits of combination therapy approaches. METHODS An extensive literature review and critical analysis of clinical trial data were conducted to inform this synthesis. RESULTS The review reveals that immunotherapy strategies, encompassing immune checkpoint inhibitors, CAR-T cell therapy, tumor vaccines, and immunomodulators, are demonstrating efficacy in the treatment of thyroid cancer. Notably, checkpoint inhibitors have been particularly effective in anaplastic and poorly differentiated thyroid cancers, albeit with challenges such as treatment resistance and adverse effects. The application of CAR-T cell therapy, successful in hematologic cancers, provides a novel perspective for thyroid cancer treatment, although its efficacy in solid tumors requires further study. Additionally, research into tumor vaccines and immunomodulators is advancing, with preliminary evidence suggesting their therapeutic potential for thyroid cancer patients. CONCLUSION The recognition of the immune microenvironment's role in treatment responsiveness is pivotal for enhancing the care of thyroid cancer patients. This review underscores the significance of combination therapy as a means to optimize treatment outcomes and charts a course for future research endeavors to broaden the spectrum of effective treatment options available to thyroid cancer patients.
Collapse
Affiliation(s)
- Ping Song
- Department of Surgical Oncology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang, China
| | - Gang Pan
- Department of Surgical Oncology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang, China
| | - Yu Zhang
- Department of Surgical Oncology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang, China
| | - Yeqin Ni
- Department of Surgical Oncology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang, China
| | - Qianyu Wang
- The Fourth Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jingjng Shi
- Department of Surgical Oncology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang, China
| | - You Peng
- Department of Surgical Oncology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang, China
| | - Ruirui Jing
- Department of Translational Medicine and Clinical Research, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Dingcun Luo
- Department of Surgical Oncology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang, China; The Fourth Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; College of Mathematical Medicine, Zhejiang Normal University, Jinhua, Zhejiang, China.
| |
Collapse
|
106
|
Li Z, Chen L, Zhang G, Wang S, Xu E, Teng J, Xu J, Peng F, Min Q, Wang Z, Shao S, Zhao L, Shan B, Wang Y, Zhan Q, Liu X. Loss of MNX1 Sensitizes Tumors to Cytotoxic T Cells by Degradation of PD-L1 mRNA. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2403077. [PMID: 39912421 PMCID: PMC11947991 DOI: 10.1002/advs.202403077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 12/04/2024] [Indexed: 02/07/2025]
Abstract
Immune checkpoint blockade (ICB) therapy, targeting programmed cell death ligand-1 (PD-L1)/programmed cell death protein 1 (PD-1) axis and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), has exhibited amazing clinical outcomes in various types of cancers. However, only a small portion of patients benefit from ICB therapy, indicating that the mechanism underlying immune checkpoint is still unclear. Here, it is reported that motor neuron and pancreas homeobox 1 (MNX1), a homeobox domain-containing transcription factor, contributes to the tumor immune escape. MNX1 increases PD-L1 expression in cancer cells by stabilizing PD-L1 mRNA rather than activating transcription. Mechanistically, MNX1 exists in the cytoplasm of cancer cells and interacts with Y-box binding protein 1 (YBX1), a multifunctional DNA/RNA-binding protein, to enhance the binding of YBX1 to PD-L1 mRNA. MNX1 ablation activates cytotoxic T cell-mediated anti-tumor immunity and sensitizes CTLA-4 blockade therapy. Moreover, MNX1 also facilitates tumor progression in an immune-independent manner in cancer cells. In addition, MNX1 is upregulated by its adjacent long non-coding RNA MNX1-AS1 via HECT and RLD domain containing E3 ubiquitin protein ligase 2 (HERC2). Together, these results reveal MNX1 as a novel immune checkpoint regulator with promising therapeutic potential.
Collapse
Affiliation(s)
- Zhengzheng Li
- Institute of Cancer Stem CellDalian Medical UniversityDalian116044China
- Soochow University Cancer InstituteSuzhou215000China
| | - Lei Chen
- Institute of Cancer Stem CellDalian Medical UniversityDalian116044China
- Department of Pulmonary OncologyAffiliated Hospital of Guangdong Medical UniversityZhanjiang524001China
| | - Ge Zhang
- Department of ImmunologyCollege of Basic Medical SciencesDalian Medical UniversityDalian116044China
| | - Shuang Wang
- Institute of Cancer Stem CellDalian Medical UniversityDalian116044China
| | - Enhang Xu
- Institute of Cancer Stem CellDalian Medical UniversityDalian116044China
| | - Jinglei Teng
- Institute of Cancer Stem CellDalian Medical UniversityDalian116044China
| | - Jiancheng Xu
- Soochow University Cancer InstituteSuzhou215000China
| | - Fang Peng
- Department of Pathologythe Second Affiliated Hospital of Dalian Medical UniversityDalian116023China
| | - Qingjie Min
- Institute of Cancer Stem CellDalian Medical UniversityDalian116044China
| | - Zhuoya Wang
- Institute of Cancer Stem CellDalian Medical UniversityDalian116044China
| | - Shujuan Shao
- University Key Laboratory of Proteomics in Liaoning ProvinceDalian Medical UniversityDalian116044China
| | - Lianmei Zhao
- Research Centerthe Fourth Hospital of Hebei Medical UniversityShijiazhuang050011China
| | - Baoen Shan
- Research Centerthe Fourth Hospital of Hebei Medical UniversityShijiazhuang050011China
| | - Yang Wang
- Institute of Cancer Stem CellDalian Medical UniversityDalian116044China
| | - Qimin Zhan
- Institute of Cancer Stem CellDalian Medical UniversityDalian116044China
- Soochow University Cancer InstituteSuzhou215000China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing)Laboratory of Molecular OncologyPeking University Cancer Hospital & InstituteBeijing100142China
| | - Xuefeng Liu
- Institute of Cancer Stem CellDalian Medical UniversityDalian116044China
| |
Collapse
|
107
|
Kshirsagar RS, Eide JG, Harris J, Abiri A, Beswick DM, Chang EH, Fung N, Hong M, Johnson BJ, Kohanski MA, Le CH, Lee JT, Nabavizadeh SA, Obermeyer IP, Pandrangi VC, Pinheiro-Neto CD, Smith TL, Snyderman CH, Suh JD, Wang EW, Wang MB, Choby G, Geltzeiler M, Lazor J, Mitchell TC, Kuan EC, Palmer JN, Adappa ND. Outcomes of Immunotherapy Treatment in Sinonasal Mucosal Melanoma. Am J Rhinol Allergy 2025; 39:102-108. [PMID: 39782303 DOI: 10.1177/19458924241308953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
BACKGROUND Sinonasal mucosal melanoma has poor survival despite multimodality treatment. While the impact of immunotherapy (IT) on metastatic cutaneous melanoma is well-defined, there are relatively little data on sinonasal mucosal melanoma. OBJECTIVE We sought to define immunotherapy outcomes in patients with sinonasal mucosal melanoma. METHODS A retrospective cohort study evaluated patients treated with IT during their overall treatment strategy for SNMM. Patient demographics, treatment, and survival outcomes were recorded. RESULTS 52 patients had IT treatment for SNMM from 2000 to 2022, with an average age of 69.1 ± 11.9 years. The most common treatment was surgery with radiation and IT (n = 26, 50%). Most regimens consisted of a combination of Nivolumab and Ipilimumab (n = 17, 32.7%) or pembrolizumab (n = 14, 26.9%). 44.2% of patients experienced reported complications. Overall survival at 1-, 2-, and 5 years was 86.9%, 74.1%, and 39.1%, respectively. CONCLUSION Approximately half of patients will have a local response following immunotherapy, but it is rare to have improvement at metastatic locations. Further research within our group will assess optimal timing and markers that are predictive of response.
Collapse
Affiliation(s)
- Rijul S Kshirsagar
- Department of Otolaryngology-Head and Neck Surgery, Kaiser Permanente Redwood City Medical Center, Redwood City, California
| | - Jacob G Eide
- Department of Otolaryngology-Head and Neck Surgery, Henry Ford Health System, Detroit, Michigan
| | - Jacob Harris
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Arash Abiri
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Daniel M Beswick
- Department of Otolaryngology-Head and Neck Surgery, University of California Los Angeles, Los Angeles, California
| | - Eugene H Chang
- Department of Otolaryngology-Head and Neck Surgery, University of Arizona, Tucson, Arizona
| | - Nicholas Fung
- Department of Otolaryngology-Head and Neck Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Michelle Hong
- Department of Otolaryngology-Head and Neck Surgery, University of California Los Angeles, Los Angeles, California
| | - Brian J Johnson
- Department of Otolaryngology-Head and Neck Surgery, Mayo Clinic, Rochester, Minnesota
| | - Michael A Kohanski
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Christopher H Le
- Department of Otolaryngology-Head and Neck Surgery, University of Arizona, Tucson, Arizona
| | - Jivianne T Lee
- Department of Otolaryngology-Head and Neck Surgery, University of California Los Angeles, Los Angeles, California
| | - Seyed A Nabavizadeh
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Isaac P Obermeyer
- Department of Otolaryngology-Head and Neck Surgery, University of California Irvine, Orange, California
| | - Vivek C Pandrangi
- Department of Otolaryngology-Head and Neck Surgery, Oregon Health & Science University, Portland, Oregon
| | | | - Timothy L Smith
- Department of Otolaryngology-Head and Neck Surgery, Oregon Health & Science University, Portland, Oregon
| | - Carl H Snyderman
- Department of Otolaryngology-Head and Neck Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jeffrey D Suh
- Department of Otolaryngology-Head and Neck Surgery, University of California Los Angeles, Los Angeles, California
| | - Eric W Wang
- Department of Otolaryngology-Head and Neck Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Marilene B Wang
- Department of Otolaryngology-Head and Neck Surgery, University of California Los Angeles, Los Angeles, California
| | - Garret Choby
- Department of Otolaryngology-Head and Neck Surgery, Mayo Clinic, Rochester, Minnesota
| | - Mathew Geltzeiler
- Department of Otolaryngology-Head and Neck Surgery, Oregon Health & Science University, Portland, Oregon
| | - Jillian Lazor
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Tara C Mitchell
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Edward C Kuan
- Department of Otolaryngology-Head and Neck Surgery, University of California Irvine, Orange, California
| | - James N Palmer
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Nithin D Adappa
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
108
|
Gu Y, Fang Y, Guo Y, Yang R, Ma J, Zhang C, Deng M, Wen Q, Gao N, Qiao H. Cytochrome P450 2E1 inhibitor Q11 is effective on hepatocellular carcinoma by promoting peritumor neutrophil chemotaxis. Int J Biol Macromol 2025; 293:139189. [PMID: 39732257 DOI: 10.1016/j.ijbiomac.2024.139189] [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: 10/24/2024] [Revised: 12/04/2024] [Accepted: 12/23/2024] [Indexed: 12/30/2024]
Abstract
Current studies found that the peritumoral tissue of hepatocellular carcinoma (HCC) may be different from normal liver tissue based on proteomics, and related to progression, recurrence and metastasis of HCC. Our previous study proposed "peritumor microenvironment (PME)" to summarize the influence of peritumor tissue on occurrence and progression of HCC. Peritumor CYP2E1 activity was significantly elevated in HCC, and related to occurrence and progression of HCC. However, the effectiveness and mechanism of inhibiting CYP2E1 against HCC remain unclear. In this study, by integrating the advantages of proteomics and transcriptomics, we reanalyzed the various influencing factors in PME. Although there were large differences in the occurrence and progression, the immunity and inflammation still played crucial roles. Peritumor neutrophil were "pro-tumor" phenotype in the stage of progression, while it showed cytotoxicity for tumor cell in the occurrence stage. CYP2E1 activity is associated with peritumor neutrophil infiltration and occurrence of HCC. CYP2E1 inhibitor Q11 showed anti-tumor effects in an orthotopic HCC mouse model by promoting secretion of chemokines and infiltration of neutrophils in peritumor tissue. Overall, these findings provided a reasonable mechanism of anti-tumor effects of CYP2E1 inhibitors, which may be a new strategy for the prevention and treatment of HCC.
Collapse
Affiliation(s)
- Yuhan Gu
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China; Department of Clinical Pharmacy, Nanyang Central Hospital, Nanyang, China
| | - Yan Fang
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yuanyuan Guo
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Rui Yang
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jun Ma
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Cunzhen Zhang
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Mengyan Deng
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Qiang Wen
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Na Gao
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Hailing Qiao
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
109
|
Moras B, Sissi C. Unravelling the Regulatory Roles of lncRNAs in Melanoma: From Mechanistic Insights to Target Selection. Int J Mol Sci 2025; 26:2126. [PMID: 40076754 PMCID: PMC11900516 DOI: 10.3390/ijms26052126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/20/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
Melanoma is the deadliest form of skin cancer, and its treatment poses significant challenges due to its aggressive nature and resistance to conventional therapies. Long non-coding RNAs (lncRNAs) represent a new frontier in the search for suitable targets to control melanoma progression and invasiveness. Indeed, lncRNAs exploit a wide range of regulatory functions along chromatin remodeling, gene transcription, post-transcription, transduction, and post-transduction to ultimately tune multiple cellular processes. The understanding of this intricate and flexible regulatory network orchestrated by lncRNAs in pathological conditions can strategically support the rational identification of promising targets, ultimately speeding up the setup of new therapeutics to integrate the currently available approaches. Here, the most recent findings on lncRNAs involved in melanoma will be analyzed. In particular, the functional links between their mechanisms of action and some frequently underestimated features, like their different subcellular localizations, will be highlighted.
Collapse
Affiliation(s)
| | - Claudia Sissi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via Marzolo 5, 35131 Padova, Italy;
| |
Collapse
|
110
|
Du F, Wang G, Dai Q, Huang J, Li J, Liu C, Du K, Tian H, Deng Q, Xie L, Zhao X, Zhang Q, Yang L, Li Y, Wu Z, Zhang Z. Targeting novel regulated cell death: disulfidptosis in cancer immunotherapy with immune checkpoint inhibitors. Biomark Res 2025; 13:35. [PMID: 40012016 DOI: 10.1186/s40364-025-00748-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 02/11/2025] [Indexed: 02/28/2025] Open
Abstract
The battle against cancer has evolved over centuries, from the early stages of surgical resection to contemporary treatments including chemotherapy, radiation, targeted therapies, and immunotherapies. Despite significant advances in cancer treatment over recent decades, these therapies remain limited by various challenges. Immune checkpoint inhibitors (ICIs), a cornerstone of tumor immunotherapy, have emerged as one of the most promising advancements in cancer treatment. Although ICIs, such as CTLA-4 and PD-1/PD-L1 inhibitors, have demonstrated clinical efficacy, their therapeutic impact remains suboptimal due to patient-specific variability and tumor immune resistance. Cell death is a fundamental process for maintaining tissue homeostasis and function. Recent research highlights that the combination of induced regulatory cell death (RCD) and ICIs can substantially enhance anti-tumor responses across multiple cancer types. In cells exhibiting high levels of recombinant solute carrier family 7 member 11 (SLC7A11) protein, glucose deprivation triggers a programmed cell death (PCD) pathway characterized by disulfide bond formation and REDOX (reduction-oxidation) reactions, termed "disulfidptosis." Studies suggest that disulfidptosis plays a critical role in the therapeutic efficacy of SLC7A11high cancers. Therefore, to investigate the potential synergy between disulfidptosis and ICIs, this study will explore the mechanisms of both processes in tumor progression, with the goal of enhancing the anti-tumor immune response of ICIs by targeting the intracellular disulfidptosis pathway.
Collapse
Affiliation(s)
- Fei Du
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China.
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Guojun Wang
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Qian Dai
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Jiang Huang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Pharmacy, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Junxin Li
- Department of pharmacy, Zigong Fourth People's Hospital, Zigong, 643000, China
| | - Congxing Liu
- Department of Pharmacy, Chengfei Hospital, Chengdu, 610000, China
| | - Ke Du
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Pediatrics, Luzhou Maternal and Child Health Hospital, Luzhou Second People's Hospital, Luzhou, 646000, Sichuan, China
| | - Hua Tian
- School of Nursing, Chongqing College of Humanities, Science & Technology, Chongqing, 401520, China
| | - Qiwei Deng
- Heruida Pharmaceutical Co.,ltd, Haikou, Hainan, 570100, China
| | - Longxiang Xie
- The TCM Hospital of Longquanyi District, Chengdu, 610100, Sichuan, China
| | - Xin Zhao
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Qimin Zhang
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Lan Yang
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Yaling Li
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Zhigui Wu
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Zhuo Zhang
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China.
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
111
|
Ascierto PA, Tang H, Dolfi S, Nyakas M, Marie Svane I, Muñoz-Couselo E, Grob JJ, Gomez-Roca CA, Chiarion-Sileni V, Peltola K, Larkin J, Melero I, Callahan M, Dummer R, Djidel P, Warad D, Reusser-Wolf D, Lipson EJ, Garnett-Benson C. Effect of prior and first-line immunotherapy on baseline immune biomarkers and modulation of the tumor microenvironment in response to nivolumab and relatlimab combination therapy in patients with melanoma from RELATIVITY-020. J Immunother Cancer 2025; 13:e009773. [PMID: 40010775 PMCID: PMC12083276 DOI: 10.1136/jitc-2024-009773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 12/02/2024] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND Some patients with melanoma experience disease progression during immunotherapy (IO) and may benefit from novel combinations of immune checkpoint inhibitors (ICIs). We report results from exploratory biomarker analyses to characterize the responses of patients with advanced melanoma to treatment with nivolumab (anti-programmed cell death-1 (PD-1)) and relatlimab (anti-lymphocyte-activation gene 3 (LAG-3)) combination therapy in RELATIVITY-020 (NCT01968109). METHODS Tumor biopsies collected at baseline and ≤4 weeks after treatment initiation were evaluated for % LAG-3-positive and % CD8-positive immune cells and % programmed death-ligand 1 (PD-L1) expression on tumor cells. Baseline biomarker expression was compared among patients with IO-refractory melanoma based on last prior therapy and IO-resistance type, and between patients with IO-refractory and IO-naïve melanoma. Change in biomarker expression after treatment was evaluated in patients with IO-refractory and IO-naïve melanoma. Immune-related gene expression was compared among resistance groups and by the last prior treatment. RESULTS Among patients with IO-refractory melanoma (N=505), elevated baseline LAG-3, PD-L1, and CD8 expression (p≤0.01, p≤0.05, p≤0.001, respectively) was observed in patients whose last prior therapy was IO versus non-IO, and in those who responded (complete/partial per Response Evaluation Criteria in Solid Tumors V.1.1) to nivolumab and relatlimab combination therapy versus those who did not (stable/progressive disease). Inflammation-related gene expression was significantly higher (p<0.05) in patients with secondary versus primary resistance to prior IO treatment, and in those whose last prior therapy was IO versus non-IO. IO-refractory patients whose tumors responded to nivolumab and relatlimab combination therapy had higher inflammation-related gene expression than non-responders (p<0.05); proliferation and hypoxia-related gene expression were enriched in non-responders. During treatment with nivolumab and relatlimab combination therapy, LAG-3 expression increased significantly in patients with IO-refractory (p≤0.01) and IO-naïve melanoma (p≤0.001), and PD-L1 and CD8 increased significantly (p≤0.01 and p≤0.05, respectively) in patients with IO-naïve melanoma. CONCLUSIONS Nivolumab and relatlimab combination therapy can modulate the tumor microenvironment in patients with both IO-refractory and IO-naïve melanoma. Further research is needed to identify patients who will most benefit from anti-LAG-3/PD-(L)1 agents, and to elucidate the mechanisms of action of, and resistance to, this combination therapy in patients with advanced melanoma. TRIAL REGISTRATION NUMBER NCT01968109.
Collapse
Affiliation(s)
- Paolo A Ascierto
- Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy
| | - Hao Tang
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Sonia Dolfi
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Marta Nyakas
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Inge Marie Svane
- Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Eva Muñoz-Couselo
- Oncology Department, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | | | - Carlos Alberto Gomez-Roca
- Department of Medicine & Clinical Research Unit, Institut Universitaire du Cancer de Toulouse Oncopole CHU Toulouse, Toulouse, France
| | - Vanna Chiarion-Sileni
- Clinical Oncology, Veneto Institute of Oncology Institute for Hospitalization and Care Scientific, Padova, Italy
| | - Katriina Peltola
- Helsinki University Central Hospital Department of Oncology, Helsinki, Finland
| | - James Larkin
- The Royal Marsden NHS Foundation Trust and The Institute of Cancer Research, London, UK
| | - Ignacio Melero
- Departments of Immunotherapy and Oncology, Clínica Universidad de Navarra, Pamplona, Navarre, Spain
| | - Margaret Callahan
- Immunotherapeutics Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Reinhard Dummer
- Department of Dermatology, University of Zürich, Zuerich, Switzerland
| | | | - Deepti Warad
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | | | - Evan J Lipson
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, Maryland, USA
| | | |
Collapse
|
112
|
Tasis A, Spyropoulos T, Mitroulis I. The Emerging Role of CD8 + T Cells in Shaping Treatment Outcomes of Patients with MDS and AML. Cancers (Basel) 2025; 17:749. [PMID: 40075597 PMCID: PMC11898900 DOI: 10.3390/cancers17050749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/13/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
CD8+ T cells are critical players in anti-tumor immunity against solid tumors, targeted by immunotherapies. Emerging evidence suggests that CD8+ T cells also play a crucial role in anti-tumor responses and determining treatment outcomes in hematologic malignancies like myelodysplastic neoplasms (MDS) and acute myeloid leukemia (AML). In this review, we focus on the implication of CD8+ T cells in the treatment response of patients with MDS and AML. First, we review reported studies of aberrant functionality and clonality of CD8+ T cells in MDS and AML, often driven by the immunosuppressive bone marrow microenvironment, which can hinder effective antitumor immunity. Additionally, we discuss the potential use of CD8+ T cell subpopulations, including memory and senescent-like subsets, as predictive biomarkers for treatment response to a variety of treatment regimens, such as hypomethylating agents, which is the standard of care for patients with higher-risk MDS, and chemotherapy which is the main treatment of patients with AML. Understanding the multifaceted role of CD8+ T cells and their interaction with malignant cells in MDS and AML will provide useful insights into their potential as prognostic/predictive biomarkers, but also uncover alternative approaches to novel treatment strategies that could reshape the therapeutic landscape, thus improving treatment efficacy, aiding in overcoming treatment resistance and improving patient survival in these challenging myeloid neoplasms.
Collapse
Affiliation(s)
- Athanasios Tasis
- Translational Research and Laboratory Medicine Unit, First Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
- Department of Hematology, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
| | - Theodoros Spyropoulos
- Department of Hematology, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
| | - Ioannis Mitroulis
- Translational Research and Laboratory Medicine Unit, First Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
- Department of Hematology, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
| |
Collapse
|
113
|
Connor C, Carr QL, Sweazy A, McMasters K, Hao H. Clinical Approaches for the Management of Skin Cancer: A Review of Current Progress in Diagnosis, Treatment, and Prognosis for Patients with Melanoma. Cancers (Basel) 2025; 17:707. [PMID: 40002300 PMCID: PMC11853469 DOI: 10.3390/cancers17040707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/16/2025] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
Melanoma represents a significant public health challenge due to its increasing incidence and potential for metastasis. This review will explore the current clinical approaches to the management of melanoma, focusing on advancements in diagnosis, treatment, and prognosis. Methods for early detection and accurate staging have been enhanced by new diagnostic strategies. Treatment modalities have expanded beyond traditional surgical excision to include targeted therapy and immunotherapy. Prognostic assessment has benefited from the development of novel biomarkers and genetic profiling. This review will highlight the progress made in the multidisciplinary management of melanoma, underscoring the importance of continuous research to improve patient outcomes.
Collapse
Affiliation(s)
- Colton Connor
- School of Medicine, University of Louisville, Louisville, KY 40202, USA; (C.C.); (Q.L.C.)
| | - Quinton L. Carr
- School of Medicine, University of Louisville, Louisville, KY 40202, USA; (C.C.); (Q.L.C.)
| | - Alisa Sweazy
- The Hiram C. Polk, Jr., MD Department of Surgery, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (A.S.); (K.M.)
| | - Kelly McMasters
- The Hiram C. Polk, Jr., MD Department of Surgery, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (A.S.); (K.M.)
| | - Hongying Hao
- The Hiram C. Polk, Jr., MD Department of Surgery, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (A.S.); (K.M.)
| |
Collapse
|
114
|
Ding J, Zhao X, Long S, Sun W, Du J, Fan J, Peng X. A Dual Stimuli-Responsive Nanoimmunomodulator for Antitumor Synergy of Macrophages and T Cells. ACS NANO 2025; 19:6468-6478. [PMID: 39919169 DOI: 10.1021/acsnano.4c17285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2025]
Abstract
Only a minority of patients benefit from current T-cell-focused adaptive immunotherapies, underscoring the need to engage innate immune cells, particularly macrophages, for multilayered tumor control. However, high-efficacy therapeutics capable of orchestrating multiple immune cells remain scarce. Herein, a dual stimuli-responsive nanoimmunomodulator (6EPP@si) that caters specifically to the tumor microenvironment (TME) is presented for the antitumor synergy of macrophages and T cells. Using the functional polymer-based carrier, we co-deliver the endoplasmic reticulum (ER)-localized photosensitizer 6E and small interfering RNA targeting CD47 (siCD47) into breast tumors. Within the acidic and high-glutathione TME, 6EPP@si undergoes self-lysosome escape and nanocleavage for precise, on-demand drug release. Consequently, siCD47 released into the cytoplasm enables potent CD47 silencing, while the ER-targeted photosensitizer 6E induces immunogenic cell death through reactive oxygen species-based ER stress, triggering the release of damage-associated molecular patterns, including calreticulin surface translocation. 6EPP@si enhances macrophage phagocytosis by modulating both antiphagocytic and prophagocytic signals and also promotes antigen presentation to activate T cells. In orthotopic breast tumor and spontaneous lung metastatic tumor models, this combined approach demonstrates robust antitumor effects and effective antimetastatic immunity, offering a meaningful strategy to simultaneously activate multiple immune cells for enhancing cancer immunotherapy.
Collapse
Affiliation(s)
- Junying Ding
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| | - Xueze Zhao
- Department of Chemistry, The University of Hong Kong, Hong Kong SAR 999077, China
| | - Saran Long
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| | - Wen Sun
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian 116024, China
- Ningbo Institute of Dalian University of Technology, Ningbo 315016, China
| | - Jianjun Du
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian 116024, China
- Ningbo Institute of Dalian University of Technology, Ningbo 315016, China
| | - Jiangli Fan
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian 116024, China
- Ningbo Institute of Dalian University of Technology, Ningbo 315016, China
| | - Xiaojun Peng
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian 116024, China
- State Key Laboratory of Fine Chemicals, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, China
| |
Collapse
|
115
|
Mehta A, Motavaf M, Nebo I, Luyten S, Osei-Opare KD, Gru AA. Advancements in Melanoma Treatment: A Review of PD-1 Inhibitors, T-VEC, mRNA Vaccines, and Tumor-Infiltrating Lymphocyte Therapy in an Evolving Landscape of Immunotherapy. J Clin Med 2025; 14:1200. [PMID: 40004731 PMCID: PMC11856346 DOI: 10.3390/jcm14041200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/08/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Melanoma, an aggressive skin cancer, presents significant therapeutic challenges. Consequently, innovative treatment strategies beyond conventional chemotherapy, radiation, and surgery are actively explored. This review discusses the evolution of immunotherapy in advanced melanoma, highlighting PD-1/PD-L1 inhibitors, mRNA vaccines, Talimogene Laherparepvec (T-VEC), and tumor-infiltrating lymphocyte (TIL) therapies. PD-1/PD-L1 inhibitors such as pembrolizumab and nivolumab block immune checkpoints, promoting T-cell cytotoxic activity and improving overall survival in patients with advanced melanoma. T-VEC, a modified oncolytic herpes virus, promotes a systemic anti-tumor response while simultaneously lysing malignant cells. mRNA vaccines, such as Moderna's mRNA-4157/V940, take advantage of malignant-cell-specific neoantigens to amplify the adaptive immune response while protecting healthy tissue. TIL therapy is a form of therapy involving ex vivo expansion and reinfusion of the patient's tumor-specific lymphocytes and has been shown to provide durable tumor control. While these therapies have demonstrated promising clinical outcomes, challenges such as tumor resistance, high financial burden, and limited accessibility pose challenges to their widespread use. This review explores combination therapies such as PD-L1 inhibitors with mRNA vaccines, or TIL therapy, which aim to enhance treatment through synergistic approaches. Further research is required to optimize these combinations, address barriers preventing their use, and control adverse events.
Collapse
Affiliation(s)
- Apoorva Mehta
- Columbia University Vagelos College of Physicians and Surgeons, 630 W 168th St, New York, NY 10032, USA; (I.N.); (S.L.); (K.D.O.-O.)
| | - Mateen Motavaf
- Duke University School of Medicine, Durham, NC 27710, USA;
| | - Ikenna Nebo
- Columbia University Vagelos College of Physicians and Surgeons, 630 W 168th St, New York, NY 10032, USA; (I.N.); (S.L.); (K.D.O.-O.)
| | - Sophia Luyten
- Columbia University Vagelos College of Physicians and Surgeons, 630 W 168th St, New York, NY 10032, USA; (I.N.); (S.L.); (K.D.O.-O.)
| | - Kofi D. Osei-Opare
- Columbia University Vagelos College of Physicians and Surgeons, 630 W 168th St, New York, NY 10032, USA; (I.N.); (S.L.); (K.D.O.-O.)
| | - Alejandro A. Gru
- Department of Dermatology, Columbia University Irving Medical Center, New York, NY 10032, USA;
| |
Collapse
|
116
|
Riaz F, Vaughn JL, Zhu H, Dickerson JC, Sayegh HE, Brongiel S, Baldwin E, Kier MW, Zaemes J, Hearn C, Abdelghany O, Cohen RB, Parikh RB, Reuss JE, Prsic E, Doroshow DB. Inpatient Immunotherapy Outcomes Study: A Multicenter Retrospective Analysis. JCO Oncol Pract 2025:OP2400788. [PMID: 39937997 DOI: 10.1200/op-24-00788] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/13/2024] [Accepted: 01/09/2025] [Indexed: 02/14/2025] Open
Abstract
PURPOSE Immune checkpoint inhibitors (ICIs) have revolutionized the care of patients with cancer, but use among hospitalized patients is controversial as a result of questionable benefit and high costs. To evaluate the role of ICIs in the inpatient (IP) setting, we conducted the Inpatient Immunotherapy Outcomes Study (IIOS) to describe characteristics and outcomes of patients who received IP ICIs. METHODS IIOS is a retrospective study of patients treated with ICIs during hospitalization between 2012 and 2021 at five academic institutions. Data collection was performed using each institution's electronic medical record. We estimated overall survival (OS) from the first administration of ICI using the Kaplan-Meier method and used adjusted Cox proportional hazards models to explore associations between clinicodemographic variables and OS. RESULTS Two hundred fifteen patients received IP ICIs (median age 60 years; 55% White; 14% Black; 13% Hispanic). Thoracic and head and neck (24%), GI (21%), and hematologic (19%) malignancies were most common. Most of the patients were ICI-naïve (75%), had stage IV solid malignancies (75%) at the time of IP ICI initiation, and had no radiographic response to ICI therapy (88%). Median OS from the first IP ICI dose was 1.55 months (95% CI, 1.08 to 1.81) for all patients and 1.28 months (95% CI, 0.95 to 1.80) for patients with advanced solid malignancies. Multivariable Cox proportional hazards model analysis found no clinicodemographic variables associated with improved OS after IP ICI administration. CONCLUSION IIOS is the largest multi-institutional effort to describe outcomes after IP ICI administration. Clinical outcomes are poor after IP ICI use and IP ICIs should be used with caution.
Collapse
Affiliation(s)
- Fauzia Riaz
- Stanford University School of Medicine, Stanford, CA
| | - John L Vaughn
- Perlmutter Cancer Center, NYU Langone Health, New York, NY
| | - Huili Zhu
- Section of Hematology and Oncology, Department of Medicine, Baylor College of Medicine, Houston, TX
| | | | - Hoda E Sayegh
- Stanford University School of Medicine, Stanford, CA
| | - Samantha Brongiel
- Smilow Cancer Hospital, Yale New Haven Health, Department of Pharmacy, New Haven, CT
| | - Elena Baldwin
- Icahn School of Medicine at Mount Sinai, New York, NY
| | | | - Jacob Zaemes
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Caleb Hearn
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Osama Abdelghany
- Smilow Cancer Hospital, Yale New Haven Health, Department of Pharmacy, New Haven, CT
| | - Roger B Cohen
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Ravi B Parikh
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Joshua E Reuss
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | | | | |
Collapse
|
117
|
Li Y, Yu Y, Lv K, Ge R, Xie X. Prognostic value of body adipose tissue parameters in cancer patients treated with immune checkpoint inhibitors. Front Immunol 2025; 16:1557726. [PMID: 40013137 PMCID: PMC11861556 DOI: 10.3389/fimmu.2025.1557726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 01/24/2025] [Indexed: 02/28/2025] Open
Abstract
Objective This study aims to explore the relationship between body adipose tissue characteristics and clinical outcomes in cancer patients receiving immune checkpoint inhibitor (ICI) therapy. Methods We conducted an extensive literature search across three major online databases-Embase, PubMed, and the Cochrane Library-to identify studies examining the link between body adipose tissue and treatment outcomes in cancer patients undergoing ICI therapy, from the inception of each database until February 20, 2024. The quality of the included studies was evaluated using the Newcastle-Ottawa Scale. The primary outcomes analyzed were hazard ratios (HRs) for overall survival (OS) and progression-free survival (PFS), as well as odds ratios (ORs) for disease control rate (DCR). Pooled estimates and 95% confidence intervals (CIs) were calculated. Results A total of 23 studies were included, encompassing 2741 cancer patients. The analysis revealed that patients with higher levels of visceral adipose tissue (VAT) exhibited significantly improved OS (HR: 0.72, 95% CI: 0.59-0.89, p < 0.001) and PFS (HR: 0.80, 95% CI: 0.67-0.96, p = 0.015), along with a higher DCR (OR: 1.81, 95% CI: 1.26-2.60, p = 0.001), compared to those with lower VAT levels. Additionally, increased subcutaneous adipose tissue (SAT) levels were associated with significantly better OS (HR: 0.69, 95% CI: 0.58-0.82, p < 0.001) and PFS (HR: 0.82, 95% CI: 0.68-1.00, p = 0.049), and a higher DCR (OR: 1.99, 95% CI: 1.15-3.44, p = 0.014). Elevated total adipose tissue (TAT) levels were also linked to longer OS (HR: 0.73, 95% CI: 0.55-0.97, p = 0.028). However, a higher visceral-to-subcutaneous adipose tissue ratio (VSR) was associated with a shorter OS (HR: 1.43, 95% CI: 1.09-1.87, p = 0.010). No significant relationship was found between TAT (HR: 0.81, 95% CI: 0.54-1.23, p = 0.332) and VSR (HR: 1.20, 95% CI: 0.95-1.51, p = 0.131) with PFS in ICI-treated patients. Conclusion This study highlights the prognostic relevance of VAT and SAT in predicting treatment response and survival outcomes in cancer patients receiving ICIs. These findings suggest that assessments of VAT and SAT should be incorporated into prognostic evaluations for this patient population.
Collapse
Affiliation(s)
- Yan Li
- Department of Traditional Chinese Medicine, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, China
| | - Yean Yu
- Department of Nephrology, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, China
| | - Kun Lv
- Department of Traditional Chinese Medicine, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, China
| | - Rongjuan Ge
- Department of Traditional Chinese Medicine, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, China
| | - Xie Xie
- Department of Traditional Chinese Medicine, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
118
|
Zielińska MK, Ciążyńska M, Sulejczak D, Rutkowski P, Czarnecka AM. Mechanisms of Resistance to Anti-PD-1 Immunotherapy in Melanoma and Strategies to Overcome It. Biomolecules 2025; 15:269. [PMID: 40001572 PMCID: PMC11853485 DOI: 10.3390/biom15020269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/22/2024] [Accepted: 01/10/2025] [Indexed: 02/27/2025] Open
Abstract
Resistance to anti-PD-1 therapy in melanoma remains a major obstacle in achieving effective and durable treatment outcomes, highlighting the need to understand and address the underlying mechanisms. The first key factor is innate anti-PD-1 resistance signature (IPRES), an expression of a group of genes associated with tumor plasticity and immune evasion. IPRES promotes epithelial-to-mesenchymal transition (EMT), increasing melanoma cells' invasiveness and survival. Overexpressed AXL, TWIST2, and WNT5a induce phenotypic changes. The upregulation of pro-inflammatory cytokines frequently coincides with EMT-related changes, further promoting a resistant and aggressive tumor phenotype. Inflamed tumor microenvironment may also drive the expression of resistance. The complexity of immune resistance development suggests that combination therapies are necessary to overcome it. Furthermore, targeting epigenetic regulation and exploring novel approaches such as miR-146a modulation may provide new strategies to counter resistance in melanoma.
Collapse
Affiliation(s)
- Magdalena K. Zielińska
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (M.K.Z.); (P.R.)
- Faculty of Medicine, Warsaw Medical University, 02-091 Warsaw, Poland
| | - Magdalena Ciążyńska
- Chemotherapy Unit and Day Chemotherapy Ward, Specialised Oncology Hospital, 97-200 Tomaszów Mazowiecki, Poland;
- Department of Dermatology, Paediatric Dermatology and Oncology Clinic, Medical University of Lodz, 91-347 Łódź, Poland
| | - Dorota Sulejczak
- Department of Experimental Pharmacology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland;
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (M.K.Z.); (P.R.)
| | - Anna M. Czarnecka
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (M.K.Z.); (P.R.)
- Department of Experimental Pharmacology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland;
| |
Collapse
|
119
|
Li WJ, Najdawi W, Badla O, Galor A, Karp CL. Immune Checkpoint Inhibitors in the Treatment of Ocular Surface Cancers: A Review. Semin Ophthalmol 2025:1-11. [PMID: 39923258 DOI: 10.1080/08820538.2025.2458658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 01/01/2025] [Accepted: 01/22/2025] [Indexed: 02/11/2025]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have transformed cancer therapy by targeting key immune pathways such as PD-1, PD-L1, CTLA-4, and LAG-3 to enhance the immune system's ability to combat malignancies. Their use in treating ocular surface tumors is an emerging area of interest, particularly in conjunctival melanoma (CM) and ocular surface squamous neoplasia (OSSN). Some studies have indicated the potential of ICI's in sebaceous gland carcinoma (SeC), conjunctival lymphoma, and Kaposi sarcoma. PURPOSE This review aims to evaluate the role of ICIs in treating ocular surface tumors, focusing on their mechanisms of action, clinical outcomes, and therapeutic potential. METHODS A literature review was conducted by searching Pubmed for studies published between January 2014 and October 2024. Studies included were original research, clinical trials, case reports and series, and reviews. RESULTS ICIs, including pembrolizumab and nivolumab, have shown promising results in CM, achieving tumor regression and disease stabilization in advanced and metastatic cases. ICIs have also demonstrated efficacy in OSSN, particularly in lesions with high tumor mutational burden, with responses ranging from partial to complete resolution. Although clinical data for SeC and conjunctival lymphoma remain limited to isolated reports, these studies suggest a role for ICIs in managing refractory or advanced disease. CONCLUSION ICIs hold transformative potential in improving outcomes for ocular surface malignancies, particularly in cases where conventional treatments fail or pose significant morbidity. Despite their promise, challenges persist, including variable response rates, immune-related adverse events, and the need for reliable predictive biomarkers. Comprehensive prospective studies are necessary to refine the application of ICIs, optimize treatment strategies, and expand therapeutic options for these challenging cancers.
Collapse
Affiliation(s)
- Wendy J Li
- Department of Ophthalmology, Bascom Palmer Eye Institute, Miami, FL, USA
| | - Wisam Najdawi
- Department of Ophthalmology, Bascom Palmer Eye Institute, Miami, FL, USA
| | - Omar Badla
- Department of Ophthalmology, Bascom Palmer Eye Institute, Miami, FL, USA
| | - Anat Galor
- Department of Ophthalmology, Bascom Palmer Eye Institute, Miami, FL, USA
- Department of Ophthalmology, Miami Veterans Hospital, Miami, FL, USA
| | - Carol L Karp
- Department of Ophthalmology, Bascom Palmer Eye Institute, Miami, FL, USA
| |
Collapse
|
120
|
Bao Y, Cruz G, Zhang Y, Qiao Y, Mannan R, Hu J, Yang F, Gondal M, Shahine M, Kang S, Mahapatra S, Chu A, Choi JE, Yu J, Lin H, Miner SJ, Robinson DR, Wu YM, Zheng Y, Cao X, Su F, Wang R, Hosseini N, Cieslik M, Kryczek I, Vaishampayan U, Zou W, Chinnaiyan AM. The UBA1-STUB1 Axis Mediates Cancer Immune Escape and Resistance to Checkpoint Blockade. Cancer Discov 2025; 15:363-381. [PMID: 39540840 PMCID: PMC11803397 DOI: 10.1158/2159-8290.cd-24-0435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 09/12/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024]
Abstract
SIGNIFICANCE Our study reveals UBA1 as a predictive biomarker for clinical outcomes in ICB cohorts, mediating cancer immune evasion and ICB resistance. We further highlight JAK1 stabilization as a key mechanism of UBA1 inhibition and nominate the UBA1-STUB1 axis as an immuno-oncology therapeutic target to improve the efficacy of ICB.
Collapse
Affiliation(s)
- Yi Bao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Gabriel Cruz
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Yuping Zhang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Yuanyuan Qiao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Rahul Mannan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Jing Hu
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Fan Yang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Mahnoor Gondal
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - Miriam Shahine
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
| | - Sarah Kang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
| | - Somnath Mahapatra
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Alec Chu
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - Jae Eun Choi
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Jiali Yu
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, Michigan
| | - Heng Lin
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, Michigan
| | - Stephanie J. Miner
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Dan R. Robinson
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Yi-Mi Wu
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Yang Zheng
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Xuhong Cao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Fengyun Su
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Rui Wang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Noshad Hosseini
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Marcin Cieslik
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - Ilona Kryczek
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, Michigan
| | - Ulka Vaishampayan
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Weiping Zou
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, Michigan
| | - Arul M. Chinnaiyan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
- Howard Hughes Medical Institute, University of Michigan, Ann Arbor, Michigan
- Department of Urology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
121
|
Wang S, Hu P, Zhang X, Fan J, Zou J, Hong W, Huang X, Pan D, Chen H, Ju D, Zhu YZ, Ye L. Recombinant CD80 fusion protein combined with discoidin domain receptor 1 inhibitor for cancer treatment. Appl Microbiol Biotechnol 2025; 109:39. [PMID: 39918582 PMCID: PMC11805834 DOI: 10.1007/s00253-025-13419-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 11/14/2024] [Accepted: 01/22/2025] [Indexed: 02/11/2025]
Abstract
Immune checkpoint inhibitors (ICIs) have significantly advanced the field of cancer immunotherapy. However, clinical data has shown that many patients have a low response rate or even resistance to immune checkpoint inhibitor alone. The underlying reasons for its poor efficacy include the deficiency of immune infiltration and effective CD28/CD80 costimulatory signal in tumor. Discoidin domain receptor 1 (DDR1) has been reported to be negatively related to immune cell infiltration in tumors. Herein, we constructed a soluble fusion protein using CD80, the natural ligand of CD28, in combination with DDR1 inhibitor. Our results demonstrated that CD80-Fc effectively activated T cells and inhibited tumor growth in vivo, even in tumors with poor efficacy of ICIs. Importantly, CD80-Fc fusion protein had a milder affinity against the targets which suggested a potential higher safety than CD28 agonists. Further, in order to promote tumor immune infiltration, we attempted to combine CD80-Fc fusion protein with DDR1 inhibitor for treatment. Our results indicated that using CD80-Fc fusion protein along with DDR1 inhibitor significantly promoted T cell infiltration in tumor microenvironment and more strongly inhibited tumor growth. Therefore, the combination use of CD80 fusion protein and DDR1 inhibitor could become an effective tumor immunotherapy strategy, potentially benefiting a larger number of patients. KEY POINTS: • We successfully constructed, expressed, and purified the recombinant CD80-Fc fusion protein • We demonstrated that CD80-Fc fusion protein has good safety and anti-tumor activity • We demonstrated that using CD80-Fc fusion protein along with DDR1 inhibitor can significantly promote immune infiltration of T cells in tumor microenvironment and more strongly inhibit tumor growth.
Collapse
Affiliation(s)
- Songna Wang
- School of Pharmacy and Laboratory of Drug Discovery From Natural Resources and Industrialization, Macau University of Science and Technology, Macau SAR, 999078, China
- Department of Biological Medicines at School of Pharmacy, Fudan University, Shanghai, 201100, China
| | - Pinliang Hu
- Beijing Beyond Biotechnology Co., Ltd, Room 308, C Building, NO.18 Xihuannanlu Street, BDA, Beijing, 100176, China
| | - Xuyao Zhang
- Department of Biological Medicines at School of Pharmacy, Fudan University, Shanghai, 201100, China
| | - Jiajun Fan
- Department of Biological Medicines at School of Pharmacy, Fudan University, Shanghai, 201100, China
| | - Jing Zou
- Beijing Beyond Biotechnology Co., Ltd, Room 308, C Building, NO.18 Xihuannanlu Street, BDA, Beijing, 100176, China
| | - Weidong Hong
- Beijing Beyond Biotechnology Co., Ltd, Room 308, C Building, NO.18 Xihuannanlu Street, BDA, Beijing, 100176, China
| | - Xuan Huang
- School of Pharmacy and Laboratory of Drug Discovery From Natural Resources and Industrialization, Macau University of Science and Technology, Macau SAR, 999078, China
- Department of Biological Medicines at School of Pharmacy, Fudan University, Shanghai, 201100, China
| | - Danjie Pan
- Department of Biological Medicines at School of Pharmacy, Fudan University, Shanghai, 201100, China
| | - Huaning Chen
- Department of Biological Medicines at School of Pharmacy, Fudan University, Shanghai, 201100, China
| | - Dianwen Ju
- Department of Biological Medicines at School of Pharmacy, Fudan University, Shanghai, 201100, China
| | - Yi Zhun Zhu
- School of Pharmacy and Laboratory of Drug Discovery From Natural Resources and Industrialization, Macau University of Science and Technology, Macau SAR, 999078, China
| | - Li Ye
- School of Pharmacy and Laboratory of Drug Discovery From Natural Resources and Industrialization, Macau University of Science and Technology, Macau SAR, 999078, China.
- Department of Biological Medicines at School of Pharmacy, Fudan University, Shanghai, 201100, China.
| |
Collapse
|
122
|
Todo M, Gatate Y, Nakano S, Kaneko G, Hagiwara M, Takahashi T, Umezawa Y, Ueda G, Ishikawa S, Makino Y, Oyama M, Shirotake S. Early detection of myocarditis caused by immune checkpoint inhibitor therapy with nivolumab and ipilimumab for advanced recurrent renal cell carcinoma. Cancer Immunol Immunother 2025; 74:97. [PMID: 39904795 PMCID: PMC11794898 DOI: 10.1007/s00262-025-03945-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 01/15/2025] [Indexed: 02/06/2025]
Abstract
Combination therapy with nivolumab and ipilimumab for advanced renal cell carcinoma (RCC) may cause immune-related myocarditis; however, its incidence in this cancer type and regimen remains unknown. At our institution, we measure biomarkers, such as high-sensitivity Troponin (hsTn), and perform electrocardiograms (ECGs) and echocardiography before and every month after the initiation of this therapy, and the findings obtained and patients' symptoms are continuously monitored by physicians and pharmacists. A retrospective survey was conducted on physiological and biochemical test findings and immune-related adverse events in patients with advanced RCC who received combination therapy with nivolumab and ipilimumab between October 1, 2018 and December 31, 2023. Patients suspected of having myocarditis consulted with cardiologists. Myocarditis due to this therapy was detected in 5 of the 86 patients (5.8%) assessed using the European Society of Cardiology 2022 guidelines. There were no fatal symptoms or death due to myocarditis. The median time to the onset of myocarditis was 25 days (21-86 days). The early detection of myocarditis caused by this therapy requires the monitoring of changes by periodically measuring hsTn and other cardiac markers and performing ECGs and echocardiography from the early stages of administration through to the end of treatment. In addition to checking symptoms, if these abnormalities are detected and myocarditis is suspected, prompt collaboration with cardiologists is recommended. Our management strategy of care by a onco-cardiology team may contribute to the early diagnosis and treatment of myocarditis.
Collapse
Affiliation(s)
- Maki Todo
- Department of Pharmacy, Saitama Medical University Saitama International Medical Center, 1397-1, Yamane, Hidaka, Saitama, 350-1298, Japan.
| | - Yodo Gatate
- Department of Cardiology, Saitama Medical University Saitama International Medical Center, Saitama, Japan
| | - Shintaro Nakano
- Department of Cardiology, Saitama Medical University Saitama International Medical Center, Saitama, Japan
| | - Go Kaneko
- Department of Uro-Oncology, Saitama Medical University Saitama International Medical Center, Saitama, Japan
| | - Masayuki Hagiwara
- Department of Uro-Oncology, Saitama Medical University Saitama International Medical Center, Saitama, Japan
| | - Takayuki Takahashi
- Department of Uro-Oncology, Saitama Medical University Saitama International Medical Center, Saitama, Japan
| | - Yuta Umezawa
- Department of Uro-Oncology, Saitama Medical University Saitama International Medical Center, Saitama, Japan
| | - Genji Ueda
- Department of Pharmacy, Saitama Medical University Saitama International Medical Center, 1397-1, Yamane, Hidaka, Saitama, 350-1298, Japan
| | - Shiho Ishikawa
- Department of Pharmacy, Saitama Medical University Saitama International Medical Center, 1397-1, Yamane, Hidaka, Saitama, 350-1298, Japan
| | - Yoshinori Makino
- Department of Pharmacy, Saitama Medical University Saitama International Medical Center, 1397-1, Yamane, Hidaka, Saitama, 350-1298, Japan
| | - Masafumi Oyama
- Department of Uro-Oncology, Saitama Medical University Saitama International Medical Center, Saitama, Japan
| | - Suguru Shirotake
- Department of Uro-Oncology, Saitama Medical University Saitama International Medical Center, Saitama, Japan
| |
Collapse
|
123
|
Yao Y, Yan Y, Suman VJ, Dietz AB, Erskine CL, Dimou A, Markovic SN, McWilliams RR, Montane HN, Block MS. Phase I study of pembrolizumab in combination with ibrutinib for the treatment of unresectable or metastatic melanoma. Front Immunol 2025; 16:1491448. [PMID: 39967670 PMCID: PMC11832643 DOI: 10.3389/fimmu.2025.1491448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 01/13/2025] [Indexed: 02/20/2025] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) have been transformative in the treatment of patients with metastatic melanoma, but primary and secondary resistance to ICI treatment is common. One key mechanism for ICI resistance is the skewing of the immune response from a cytotoxic (Th1) to a chronic inflammatory (Th2) profile. The small molecule ibrutinib is a dual-target agent that inhibits Bruton's Tyrosine Kinase (BTK) and Interleukin-2-inducible T-cell Kinase (ITK), a key regulator of Th2 immunity. Therefore, combining ibrutinib and pembrolizumab could potentially induce an increase in Th1 immune polarity in melanoma patients. We hypothesize that the combination would be well-tolerated and might result in clinical benefit for patients with metastatic melanoma. The primary aim of this phase I study was to evaluate the safety, tolerability, and determine the maximum tolerated dose (MTD) of ibrutinib in combination with pembrolizumab in patients with metastatic melanoma. Methods A 3 + 3 phase I clinical trial was conducted in patients with unresectable Stage III or metastatic melanoma (stage IV) not amenable to local therapy. Pembrolizumab (200 mg/kg every 3 weeks) was combined with ibrutinib, administered orally at the dose assigned at the time of registration (140 mg daily, 280 mg daily, and 420 mg daily). Patients were treated until disease progression, intolerability, or patient decision to discontinue. Blood samples were collected after each cycle of treatment for immunophenotyping and Th1/Th2 polarity assessment based on immune response markers. Results Between January 31, 2017 and January 9, 2023, 17 patients were enrolled. The MTD of ibrutinib in combination with pembrolizumab was determined to be 420 mg daily. The adverse events leading to discontinuation included: grade 4 ALT and AST increase (1 pt, DL0); grade 4 ALT increase with grade 3 AST increase (1 pt, DL1); and grade 3 hyponatremia, hypoxia, and maculo-papular rash (1 pt, DL1). Three of the 16 patients treated had objective responses (2 partial responses, 1 complete response) lasting over 8 months. The median progression-free survival was 3 months, and median and overall survival was 1.8 years. The combination treatment did not result in consistent increase in Th1 immune polarity. Conclusion In conclusion, the maximum tolerated dose of ibrutinib in combination with pembrolizumab in patients with advanced or metastatic melanoma was established at 420 mg by mouth once daily. The combination was well-tolerated but did not result in a consistent increase in Th1 immune polarity; further investigation is needed to assess the relative clinical efficacy of this approach. (Funded by Pharmacyclics; ClinicalTrials.gov number: NCT03021460). Clinical trial registration www.clinicaltrials.gov, identifier NCT03021460.
Collapse
Affiliation(s)
- Yuan Yao
- Department of Oncology, Mayo Clinic, Rochester, MN, United States
| | - Yiyi Yan
- Division of Hematology and Oncology, Department of Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Vera J. Suman
- Department of Health Sciences, Division of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, United States
| | - Allan B. Dietz
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | | | - Anastasios Dimou
- Department of Oncology, Mayo Clinic, Rochester, MN, United States
| | - Svetomir N. Markovic
- Department of Oncology, Mayo Clinic, Rochester, MN, United States
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
| | | | | | - Matthew S. Block
- Department of Oncology, Mayo Clinic, Rochester, MN, United States
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
124
|
Tintelnot J, Paschold L, Goekkurt E, Schultheiss C, Matschl U, Santos Cruz M, Bauer M, Wickenhauser C, Thuss-Patience P, Lorenzen S, Ettrich TJ, Riera-Knorrenschild J, Jacobasch L, Kretzschmar A, Kubicka S, Al-Batran SE, Reinacher-Schick A, Pink D, Bokemeyer C, Sinn M, Lindig U, Hinke A, Hegewisch-Becker S, Stein A, Binder M. Inflammatory Stress Determines the Need for Chemotherapy in Patients with HER2-Positive Esophagogastric Adenocarcinoma Receiving Targeted Therapy and Immunotherapy. Cancer Immunol Res 2025; 13:200-209. [PMID: 39527097 PMCID: PMC11788649 DOI: 10.1158/2326-6066.cir-24-0561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 08/10/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
Anti-PD-1, trastuzumab, and chemotherapy are used in the treatment of patients with advanced HER2-positive esophagogastric adenocarcinoma, but long-term survival remains limited. In this study, we report extended follow-up data from the INTEGA trial (NCT03409848), which investigated the efficacy of the anti-PD-1 nivolumab, trastuzumab, and FOLFOX chemotherapy (FOLFOX arm) in comparison with a chemotherapy-free regimen involving nivolumab, trastuzumab, and the anti-CTLA-4 ipilimumab (Ipi arm) in the first-line setting for advanced disease. The 12-month overall survival (OS) showed no statistical difference between the arms, with 57% OS (95% confidence interval, 41%-71%) in the Ipi arm and 70% OS (95% confidence interval, 54%-82%) in the FOLFOX arm. Crossing of the survival curves indicated a potential long-term benefit for some patients within the Ipi arm, but early progressors in the Ipi arm underlined the need for biomarker-guided strategies to optimize treatment selection. To this end, metabolomic and cytokine analyses demonstrated elevated levels of normetanephrine, cortisol, and IL6 in immunotherapy-unresponsive patients in the Ipi arm, suggesting a role for systemic inflammatory stress in modulating antitumor immune responses. Patients with this signature also showed an increased neutrophil to lymphocyte ratio that persisted in the Ipi arm, but not in the FOLFOX arm, and strongly correlated with survival. Furthermore, a low neutrophil to lymphocyte ratio characterized patients benefiting from immunotherapy and targeted therapy without the need for additional chemotherapy. These data suggest that patient selection based on inflammatory stress-driven immune changes could help customize first-line treatment in patients with advanced HER2-positive esophagogastric adenocarcinoma to potentially improve long-term survival.
Collapse
Affiliation(s)
- Joseph Tintelnot
- II. Medical Clinic and Polyclinic, University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lisa Paschold
- Department of Internal Medicine IV - Oncology/Hematology, University Hospital, Martin-Luther University, Halle, Germany
| | - Eray Goekkurt
- II. Medical Clinic and Polyclinic, University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hematology-Oncology Practice Eppendorf (HOPE), Hamburg, Germany
| | - Christoph Schultheiss
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
- Laboratory of Translational Immuno-Oncology, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
| | - Urte Matschl
- Department of Virus Immunology, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Mariana Santos Cruz
- II. Medical Clinic and Polyclinic, University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marcus Bauer
- Institute of Pathology, University Hospital, Martin-Luther University, Halle, Germany
| | - Claudia Wickenhauser
- Institute of Pathology, University Hospital, Martin-Luther University, Halle, Germany
| | | | - Sylvie Lorenzen
- Rechts der Isar Hospital, Technical University of Munich, Munich, Germany
| | | | | | | | | | | | | | | | - Daniel Pink
- Klinik und Poliklinik für Innere Medizin C, University Greifswald, Greifswald, Germany
- Klinik für Hämatologie, Onkologie und Palliativmedizin, Sarkomzentrum Berlin-Brandenburg, HELIOS Klinikum Bad-Saarow, Bad Saarow, Germany
| | - Carsten Bokemeyer
- II. Medical Clinic and Polyclinic, University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marianne Sinn
- II. Medical Clinic and Polyclinic, University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | | | | - Alexander Stein
- II. Medical Clinic and Polyclinic, University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mascha Binder
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
- Laboratory of Translational Immuno-Oncology, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
| |
Collapse
|
125
|
Lordick F, Mauer ME, Stocker G, Cella CA, Ben-Aharon I, Piessen G, Wyrwicz L, Al-Haidari G, Fleitas-Kanonnikoff T, Boige V, Lordick Obermannová R, Martens UM, Gomez-Martin C, Thuss-Patience P, Arrazubi V, Avallone A, Shiu KK, Artru P, Brenner B, Buges Sanchez C, Chau I, Lorenzen S, Daum S, Sinn M, Merelli B, van Grieken NCT, Nilsson M, Collienne M, Giraut A, Smyth E. Adjuvant immunotherapy in patients with resected gastric and oesophagogastric junction cancer following preoperative chemotherapy with high risk for recurrence (ypN+ and/or R1): European Organisation of Research and Treatment of Cancer (EORTC) 1707 VESTIGE study. Ann Oncol 2025; 36:197-207. [PMID: 39542422 DOI: 10.1016/j.annonc.2024.10.829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 10/31/2024] [Accepted: 10/31/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND Patients with gastro-oesophageal adenocarcinoma with tumour-positive lymph nodes (ypN+) or positive surgical margins (R1) following neoadjuvant chemotherapy and resection are at high risk of recurrence. Adjuvant nivolumab is effective in oesophageal/oesophagogastric junction cancer and residual pathological disease following chemoradiation and surgery. Immune checkpoint inhibition has shown efficacy in advanced gastro-oesophageal cancer. We hypothesised that nivolumab/ipilimumab would be more effective than adjuvant chemotherapy in high-risk (ypN+ and/or R1) patients with gastro-oesophageal adenocarcinoma following neoadjuvant chemotherapy and resection. PATIENTS AND METHODS VESTIGE was an academic international, multicentre, open-label, randomised phase II trial evaluating the efficacy of adjuvant nivolumab/ipilimumab versus chemotherapy in gastro-oesophageal adenocarcinoma at high risk of recurrence. Patients were randomised 1 : 1 to receive standard adjuvant chemotherapy (same regimen as neoadjuvant) or nivolumab 3 mg/kg intravenously (i.v.) every 2 weeks plus ipilimumab 1 mg/kg i.v. every 6 weeks for 1 year. Key inclusion criteria included ypN+ and/or R1 status after neoadjuvant chemotherapy plus surgery. The primary endpoint was disease-free survival in the intent-to-treat population. Secondary endpoints included overall survival, locoregional and distant failure rates, and safety according to National Cancer Institute Common Terminology Criteria for Adverse Events v5.0. RESULTS The independent Data Monitoring Committee reviewed data from 189 of the planned 240 patients in June 2022 and recommended stopping recruitment due to futility. At the time of final analysis, median follow-up was 25.3 months for 195 patients (98 nivolumab/ipilimumab and 97 chemotherapy). Median disease-free survival for the nivolumab/ipilimumab group was 11.4 months [95% confidence interval (CI) 8.4-16.8 months] versus 20.8 months (95% CI 15.0-29.9 months) for the chemotherapy group, hazard ratio 1.55 (95% CI 1.07-2.25, one-sided P = 0.99). The 12-month disease-free survival rates were 47.1% and 64.0%, respectively. There were no toxicity concerns or excess early discontinuations. CONCLUSION Nivolumab/ipilimumab did not improve disease-free survival compared with chemotherapy in patients with ypN+ and/or R1 gastro-oesophageal adenocarcinoma following neoadjuvant chemotherapy and surgery.
Collapse
Affiliation(s)
- F Lordick
- Department of Medicine (Oncology, Gastroenterology, Hepatology, Pulmonology), University of Leipzig Medical Center, Comprehensive Cancer Center Central Germany (CCCG), Leipzig, Germany.
| | - M E Mauer
- European Organisation for Research and Treatment of Cancer (EORTC) Headquarters, Brussels, Belgium
| | - G Stocker
- Department of Medicine (Oncology, Gastroenterology, Hepatology, Pulmonology), University of Leipzig Medical Center, Comprehensive Cancer Center Central Germany (CCCG), Leipzig, Germany
| | - C A Cella
- Istituto Europeo di Oncologia, IRCCS, Milan, Italy
| | - I Ben-Aharon
- Department of Oncology, Rambam Health Care Campus, Haifa, Israel
| | - G Piessen
- CHRU de Lille-Hôpital Huriez, Lille, France
| | - L Wyrwicz
- Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - G Al-Haidari
- Oslo University Hospital-Ullevaal Hospital, Oslo, Norway
| | - T Fleitas-Kanonnikoff
- Hospital Clinico Universitario De Valencia, Incliva Biomedical Research Institute, Valencia, Spain
| | - V Boige
- Department of Cancer Medicine, Institut Gustave Roussy, Villejuif, France
| | - R Lordick Obermannová
- Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - U M Martens
- Department of Oncology, SLK-Kliniken Heilbronn, Heilbronn, Germany
| | | | - P Thuss-Patience
- Charité-Universitäetsmedizin Berlin-Charité Campus Virchow-Klinikum, Berlin, Germany
| | - V Arrazubi
- Hospital Universitario de Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - A Avallone
- Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - K K Shiu
- University College Hospital, London, UK
| | - P Artru
- Hôpital Privé Jean Mermoz, Lyon, France
| | - B Brenner
- Rabin Medical Center, Beilinson Hospital, Petach Tikva, Israel
| | - C Buges Sanchez
- Department of Medical Oncology, ICO-Badalona (Catalan Institute of Oncology-Badalona), Barcelona, Spain
| | - I Chau
- Royal Marsden Hospital, London, UK
| | - S Lorenzen
- Department of Haematology and Oncology, Klinikum rechts der Isar, Technische Universitaet Muenchen, Munich, Germany
| | - S Daum
- Charité-Universitäetsmedizin Berlin-Charité Campus Benjamin-Franklin, Berlin, Germany
| | - M Sinn
- University Medical Center Hamburg-Eppendorf, Center of Oncology, Hamburg, Germany
| | - B Merelli
- Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | - N C T van Grieken
- Department of Pathology, Amsterdam UMC, location VUmc, Cancer Center Amsterdam, VU University, Amsterdam, The Netherlands
| | - M Nilsson
- Division of Surgery and Oncology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Solna, Sweden; Department of Upper Abdominal Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - M Collienne
- European Organisation for Research and Treatment of Cancer (EORTC) Headquarters, Brussels, Belgium
| | - A Giraut
- European Organisation for Research and Treatment of Cancer (EORTC) Headquarters, Brussels, Belgium
| | - E Smyth
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, UK
| |
Collapse
|
126
|
Tran S, Forrest N, Guggilla V, Perottino G, Johnson J, Sosman J, Roy I, Walunas T. Weight and Blood-Based Markers of Cachexia Predict Disability, Hospitalization and Worse Survival in Cancer Immunotherapy Patients. J Cachexia Sarcopenia Muscle 2025; 16:e13685. [PMID: 39817619 PMCID: PMC11736629 DOI: 10.1002/jcsm.13685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 08/05/2024] [Accepted: 11/18/2024] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND Cancer-associated cachexia can inhibit immune checkpoint inhibitor (ICI) therapy efficacy. Cachexia's effect on ICI therapy has not been studied in large cohorts of cancer patients aside from lung cancer. We studied associations between real-world routinely collected clinical cachexia markers and disability-free, hospitalization-free and overall survival of cancer patients. METHODS A retrospective study was conducted of electronic health records (EHR) of patients with lung, renal cell, melanoma and other cancers treated with ICI therapy at Northwestern Medicine of Chicago, IL, United States, between March 2011 and January 2022. Weight, body mass index, absolute neutrophil and lymphocyte counts, albumin and C-reactive protein (CRP) measures were analysed to calculate the Fearon consensus criteria for cachexia, weight loss grading system (WLGS) score, neutrophil-lymphocyte ratio (NLR), Prognostic Nutritional Index (PNI) and modified Glasgow Prognostic Score (mGPS) at ICI therapy initiation. Kaplan-Meier and Cox proportional hazards analyses were used to determine associations between these metrics and disability-free, hospitalization-free and overall survival. RESULTS EHR analysis uncovered 3285 cancer patients on ICI therapy (54% > 65 years of age, 50.7% male, 77.7% White). At ICI therapy initiation, 1282 (39.0%) patients had cachexia (consensus criteria), 1641 (50.0%) had a WLGS score ≥ 2, 1806 (55.0%) had an NLR > 3, 1087 (33.1%) had albumin < 3.5 g/dL and 1318 (40.1%) had a PNI < 44. Missing measurements included CRP missing for 98.2% and mGPS missing for 98.6% of patients. Disability-free (n = 1373), hospitalization-free (n = 2374) and overall survival (n = 1599) events were analysed with 1-year rates of 65% (64%-67%), 35% (34%-37%) and 65% (63%-66%), respectively. Multivariate Cox model analyses showed hazard ratios (HR) for cachexia at 1.58 (95% CI 1.38-1.80), 1.47 (95% CI 1.33-1.63) and 1.97 (95% CI 1.75-2.23) for disability, hospitalization and death, respectively. HRs for WLGS ≥ 2 were 1.45 (95% CI 1.28-1.66), 1.37 (95% CI 1.24-1.51) and 1.91 (95% CI 1.69-2.17). HRs for NLR > 3 were 1.57 (95% CI 1.35-1.83), 1.40 (95% CI 1.25-1.58) and 1.95 (95% CI 1.67-2.27). HRs for albumin < 3.5 g/dL were 1.33 (95% CI 1.15-1.54), 1.67 (95% CI 1.50-1.86) and 2.09 (95% CI 1.84-2.36). HRs for PNI < 44 were 1.60 (95% CI 1.39-1.84), 1.46 (95% CI 1.31-1.63) and 2.07 (95% CI 1.80-2.37). CONCLUSIONS Fearon consensus criteria, WLGS, NLR, albumin and PNI were routinely collected at ICI initiation in regular clinical practice and predictive of worse disability-free, hospitalization-free and overall survival in cancer patients receiving ICI therapy. These routine clinical measures may aid prognostication and decision-making in cancer patients with cachexia.
Collapse
Affiliation(s)
- Steven D. Tran
- Center for Health Information PartnershipsNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Noah J. Forrest
- Center for Health Information PartnershipsNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Vijeeth Guggilla
- Center for Health Information PartnershipsNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | | | - Jodi L. Johnson
- Robert H Lurie Comprehensive Cancer Center of Northwestern UniversityChicagoIllinoisUSA
- Departments of Pathology, Dermatology and Medical Social SciencesNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Jeffrey Sosman
- Robert H Lurie Comprehensive Cancer Center of Northwestern UniversityChicagoIllinoisUSA
- Department of Medicine, Division of OncologyNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Ishan Roy
- Robert H Lurie Comprehensive Cancer Center of Northwestern UniversityChicagoIllinoisUSA
- Shirley Ryan AbilityLabChicagoIllinoisUSA
- Department of Physical Medicine and RehabilitationNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Theresa L. Walunas
- Center for Health Information PartnershipsNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Robert H Lurie Comprehensive Cancer Center of Northwestern UniversityChicagoIllinoisUSA
- Department of Medicine, Division of General Internal MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| |
Collapse
|
127
|
Lin J, Li W, Zhang X, Zhou K, Yang Y, Cheng S, Sun R, Dang C, Diao D. Thromboembolic events associated with immune checkpoint inhibitors in cancer patients: A Bayesian network meta-analysis. Thromb Res 2025; 246:109243. [PMID: 39721224 DOI: 10.1016/j.thromres.2024.109243] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 12/17/2024] [Accepted: 12/19/2024] [Indexed: 12/28/2024]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs), which offer previously unknown therapeutic advantages, have revolutionized cancer treatment. However, the risk of thromboembolic events (TEEs) associated with ICIs remains unclear. The aim of this network meta-analysis (NMA) was to evaluate the incidence of TEEs in cancer patients receiving different treatment regimens. METHODS We searched for randomized clinical trials (RCTs) between January 2021 and December 2023 without restricting the cancer type. The percentages of TEEs were systematically extracted. An NMA was performed comparing atezolizumab, cemiplimab, durvalumab, ipilimumab, nivolumab, pembrolizumab, conventional therapy (which consists mainly of chemotherapy, targeted therapy, placebo, and their combinations), two ICI drugs, one ICI drug combined with conventional therapy, and two ICI drugs combined with conventional therapy. Additionally, subgroup analysis was conducted based on cancer type. RESULTS Eighty-three RCTs involving 54,736 patients were included. Patients receiving ICIs demonstrated comparable risks of arterial thromboembolism (ATE), deep vein thrombosis (DVT), myocardial infarction (MI), and cerebrovascular accidents (CVAs). Nivolumab (OR 0.39, 95 % CI 0.19 to 0.80) and two ICI drugs (OR 0.52, 95 % CI 0.29 to 0.89) had the lowest risk of venous thromboembolism (VTE) compared to two ICI drugs with conventional therapy. The risk of pulmonary embolism (PE) was greater for ipilimumab (OR 4.09, 95 % CI 1.13 to 15.51) than for nivolumab. For melanoma in the subgroup analysis, nivolumab significantly reduced the risk of VTE (OR 0.07, 95 % CI 0.00 to 0.76) compared to two ICI drugs. Among the single-ICI regimens, durvalumab was associated with the highest incidence of ATE, MI, and CVAs; ipilimumab had the highest incidence of VTE and PE; and pembrolizumab had the highest incidence of DVT. The combination of one ICI drug with conventional therapy was associated with a significantly greater risk of TEEs (except for MI) than the combination of two ICI drugs. CONCLUSIONS Various ICI regimens in cancer patients exhibit clinically significant differences in the risks of TEEs. Nivolumab exhibited a favorable safety profile regarding VTE, while ipilimumab had the highest risk of both VTE and PE. Different ICI regimens require tailored risk management strategies to reduce TEEs.
Collapse
Affiliation(s)
- Jinhe Lin
- Department of Oncology Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Wenxing Li
- Department of Oncology Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Xin Zhang
- Department of Oncology Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Kai Zhou
- Department of Oncology Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Yanqi Yang
- Department of Pathology, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Shaoli Cheng
- Basic Medical Experiment Teaching Center, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Ruifang Sun
- Department of Pathology, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Chengxue Dang
- Department of Oncology Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
| | - Dongmei Diao
- Department of Oncology Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
| |
Collapse
|
128
|
Eochagain CM, Neuendorff NR, Gente K, Leipe J, Verhaert M, Sam C, de Glas N, Kadambi S, Canin B, Gomes F, Decoster L, Korc-Grodzicki B, Rostoft S, Battisti NML, Wildiers H. Management of immune checkpoint inhibitor-associated toxicities in older adults with cancer: recommendations from the International Society of Geriatric Oncology (SIOG). Lancet Oncol 2025; 26:e90-e102. [PMID: 39914430 DOI: 10.1016/s1470-2045(24)00404-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 07/04/2024] [Accepted: 07/17/2024] [Indexed: 05/07/2025]
Abstract
Immune checkpoint inhibitors (ICIs) have substantially advanced the treatment landscape for a wide variety of malignancies. Older adults represent a large and rapidly growing demographic, among whom ICIs are widely prescribed. Management of ICI-associated toxicity among older adults, particularly in the presence of frailty and comorbidity, poses unique challenges. In this Policy Review, developed by the International Society of Geriatric Oncology (SIOG), we offer an evidence-based framework for health-care providers, caregivers, and policy makers for treating older adults with ICIs, focusing on unique considerations for this population that are not adequately addressed by existing guidelines, and expanding them to encompass geriatric oncology principles.
Collapse
Affiliation(s)
- Colm Mac Eochagain
- Trinity St James' Cancer Institute, Dublin, Ireland; Mercer Institute for Successful Aging, St James' Hospital, Dublin, Ireland.
| | - Nina Rosa Neuendorff
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, University Duisburg-Essen, Essen, Germany; Department of Geriatric Medicine, Marien Hospital Herne, Ruhr-Universität Bochum, Bochum, Germany
| | - Karolina Gente
- Department of Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Jan Leipe
- Division of Rheumatology, Department of Medicine V, University Medical Centre Mannheim, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany; Division of Rheumatology, Department of Internal Medicine I, University Medical Centre Schleswig-Holstein, Kiel, Germany
| | - Marthe Verhaert
- Laboratory for Medical and Molecular Oncology, Translational Oncology Research Center, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | | | - Nienke de Glas
- Department of Medical Oncology, Helse Førde, Førde, Norway; Department of Medical Oncology, Leiden University Medical Center, Leiden, Netherlands
| | - Sindhuja Kadambi
- Division of Hematology/Oncology, Department of Medicine, James P Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Beverly Canin
- Cancer and Aging Research Group, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Fabio Gomes
- Department of Medical Oncology, The Christie Hospital, Manchester, UK
| | - Lore Decoster
- Department of Medical Oncology, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | | | - Siri Rostoft
- Department of Geriatric Medicine and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | - Hans Wildiers
- Department of General Medical Oncology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
129
|
Foerster Y, Mayer K, Wasserer S, Dechant M, Verkhoturova V, Heyer S, Biedermann T, Persa O. Elevated Neutrophil-to-Lymphocyte Ratio Correlates With Liver Metastases and Poor Immunotherapy Response in Stage IV Melanoma. Cancer Med 2025; 14:e70631. [PMID: 39931836 PMCID: PMC11811709 DOI: 10.1002/cam4.70631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 01/02/2025] [Accepted: 01/16/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Immune checkpoint inhibition (ICI) has revolutionized treatment for metastasized melanoma, but many patients remain unresponsive. Concerning potential adverse events, reliable biomarkers to predict ICI response are needed. In this context, neutrophil-to-lymphocyte ratio (NLR) and derived NLR (dNLR) have emerged. Liver metastases also limit ICI efficacy, correlating with diminished overall survival (OS) and progression-free survival (PFS) and may siphon activated T cells from the systemic circulation, creating an 'immune desert state'. We evaluated the predictive role of NLR and dNLR for ICI response and the impact of liver metastases on systemic immunity and treatment efficacy. PATIENTS AND METHODS In this single-center retrospective study, we included 141 stage IV melanoma patients undergoing ICI. NLR and dNLR were calculated from absolute neutrophil count, absolute lymphocyte count, and white blood cell count. RESULTS Elevated NLR and dNLR were associated with poor response to ICI and inferior PFS. Patients with liver metastases exhibited higher NLR and dNLR levels and showed diminished response to ICI. CONCLUSIONS Elevated baseline NLR and dNLR predict poor response to ICI and PFS in stage IV melanoma. Liver metastases are negative predictors for ICI response, with associated higher NLR and dNLR levels potentially contributing to therapy resistance.
Collapse
Affiliation(s)
- Yannick Foerster
- Department of DermatologyTUM School of Medicine and HealthMünchenGermany
| | - Kristine Mayer
- Department of DermatologyTUM School of Medicine and HealthMünchenGermany
| | - Sophia Wasserer
- Department of DermatologyTUM School of Medicine and HealthMünchenGermany
| | - Marta Dechant
- Department of DermatologyTUM School of Medicine and HealthMünchenGermany
| | | | - Sarah Heyer
- Department of DermatologyTUM School of Medicine and HealthMünchenGermany
| | - Tilo Biedermann
- Department of DermatologyTUM School of Medicine and HealthMünchenGermany
| | - Oana‐Diana Persa
- Department of DermatologyTUM School of Medicine and HealthMünchenGermany
| |
Collapse
|
130
|
Höppener DJ, Grünhagen DJ, Eggermont AMM, van der Veldt AAM, Verhoef C. An Overview of Liver Directed Locoregional Therapies. Hematol Oncol Clin North Am 2025; 39:103-123. [PMID: 39510668 DOI: 10.1016/j.hoc.2024.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
An overview of all liver-directed locoregional therapies, including surgical resection for melanoma liver metastases (MLMs), is provided. MLM patients are divided by their primary melanoma location; cutaneous, uvea (eye), and mucosal melanoma. If patients with isolated cutaneous MLMs are considered for surgical resection, treatment with systemic therapy should be part of the treatment course. For uveal MLMs, complete surgical or ablative treatment of all MLMs suggests superior results compared with other liver-directed or systemic therapies, based on current evidence, no recommendations for any liver-directed regional therapy in the treatment of mucosal MLMs can be made.
Collapse
Affiliation(s)
- Diederik J Höppener
- Department of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands
| | - Dirk J Grünhagen
- Department of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands
| | - Alexander M M Eggermont
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, the Netherlands
| | - Astrid A M van der Veldt
- Department of Medical Oncology, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands
| | - Cornelis Verhoef
- Department of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands.
| |
Collapse
|
131
|
Triantafyllou E, Gudd CLC, Possamai LA. Immune-mediated liver injury from checkpoint inhibitors: mechanisms, clinical characteristics and management. Nat Rev Gastroenterol Hepatol 2025; 22:112-126. [PMID: 39663461 DOI: 10.1038/s41575-024-01019-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/05/2024] [Indexed: 12/13/2024]
Abstract
Immunotherapy has changed the treatment landscape for patients with cancer in the past decade. Immune checkpoint inhibitor (ICI)-based therapies have proven effective in a range of malignancies, including liver and gastrointestinal cancers, but they can cause diverse off-target organ toxicities. With the increasingly wider application of these drugs, immune-mediated liver injury from ICIs has become a commonly encountered challenge in clinical hepatology and gastroenterology. In this Review, we discuss the evidence from human and animal studies on the immunological mechanisms of immune-mediated liver injury from ICIs and summarize its clinical features and practical considerations for its management.
Collapse
Affiliation(s)
- Evangelos Triantafyllou
- Section of Hepatology and Gastroenterology, Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom.
| | - Cathrin L C Gudd
- Section of Hepatology and Gastroenterology, Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Lucia A Possamai
- Section of Hepatology and Gastroenterology, Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom.
- Liver and Antiviral Unit, St Mary's Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom.
| |
Collapse
|
132
|
Obozina AS, Pakhomov AA, Frolova AY, Deyev SM, Shipunova VO. Optimizing combination targeted immunotoxin therapy: Insights from HER2 and EpCAM expression profiles. Biochem Biophys Res Commun 2025; 746:151218. [PMID: 39752974 DOI: 10.1016/j.bbrc.2024.151218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 12/19/2024] [Indexed: 01/15/2025]
Abstract
Molecular targeted cancer therapy is a rapidly developing field, driving progress toward greater treatment efficacy. However, targeted monotherapy often fails due to the development of multidrug resistance in tumors. The combination of multiple targeted agents emerges as a possible solution to enhance treatment outcomes by activating different signaling pathways. This study systematically investigates the combined effect of targeted agents for the oncomarkers HER2 and EpCAM on cancer cells. Specifically, the study examined the impact of anti-HER2 (DARP_9.29-LoPE) and anti-EpCAM (DARP_EC1-LoPE) immunotoxins on a panel of cancer cells expressing various levels of HER2 and EpCAM. Using the Chou-Talalay combination indices, the study revealed that cells with low HER2 expression and high EpCAM expression are not optimal targets for combined HER2/EpCAM therapy. In contrast, the most effective approach involves the usage of an equimolar ratio of immunotoxins for cells exhibiting high HER2 and moderate EpCAM expression, resulting in a synergistic therapeutic effect. These findings provide significant insights into optimizing combination anti-HER2/EpCAM therapies and hold promise for the development of more effective cancer treatment strategies.
Collapse
Affiliation(s)
- A S Obozina
- Moscow Center for Advanced Studies, Kulakova Str. 20, 123592, Moscow, Russia
| | - A A Pakhomov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya St., 117997, Moscow, Russia
| | - A Yu Frolova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya St., 117997, Moscow, Russia
| | - S M Deyev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya St., 117997, Moscow, Russia
| | - V O Shipunova
- Moscow Center for Advanced Studies, Kulakova Str. 20, 123592, Moscow, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya St., 117997, Moscow, Russia.
| |
Collapse
|
133
|
Kreft S, Lorigan P. Combination Immunotherapy for Advanced Melanoma-How to Choose? J Clin Oncol 2025; 43:478-479. [PMID: 39496095 DOI: 10.1200/jco-24-02005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 10/10/2024] [Indexed: 11/06/2024] Open
Affiliation(s)
- Sophia Kreft
- Sophia Kreft, MB, The Christie Hospital NHS Foundation Trust, Manchester, United Kingdom; and Paul Lorigan, MB, BCH, The Christie Hospital NHS Foundation Trust, Manchester, United Kingdom, Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Paul Lorigan
- Sophia Kreft, MB, The Christie Hospital NHS Foundation Trust, Manchester, United Kingdom; and Paul Lorigan, MB, BCH, The Christie Hospital NHS Foundation Trust, Manchester, United Kingdom, Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
134
|
Hung Y, Lee P, Chang Y, Yang M, Chiu C, Chen M, Lan K, Lee I, Hou M, Chao Y, Huang Y. Hepatic Events During Immune Checkpoint Inhibitor Treatment Between Liver and Non-Liver Malignancies in Hepatitis B Endemic Areas. Aliment Pharmacol Ther 2025; 61:501-512. [PMID: 39582238 PMCID: PMC11707645 DOI: 10.1111/apt.18403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 07/29/2024] [Accepted: 11/07/2024] [Indexed: 11/26/2024]
Abstract
BACKGROUND Notable advances have been made in immune checkpoint inhibitors (ICIs) for cancer treatment. However, the adverse effects of ICIs, especially hepatotoxicity, remain a challenging problem. Whether patients in hepatitis B virus (HBV)-endemic areas are prone to developing hepatic adverse events during ICI treatment warrants further exploration. METHODS From 2014 to 2020, the data of all patients with cancer who received ICI treatment at Taipei Veterans General Hospital were retrospectively reviewed. The incidence of and risk factors for hepatic adverse events, including hepatitis flare, immune-related hepatitis (irHepatitis) and HBV reactivation (HBVr), were analysed through a Cox proportional hazard regression model. RESULTS A total of 1283 patients with cancer (190 hepatocellular carcinoma [HCC] patients and 1093 patients with non-HCC malignancies) were eligible for analysis, of whom 283 (22.1%) were HBsAg-positive. The incidence of hepatitis flare events of any grade was significantly higher in HCC patients than in non-HCC patients (45.8% vs. 25.6%, p < 0.001). HCC and baseline alanine aminotransferase (ALT) > 40 U/L were independent risk factors for ≥ grade 3 hepatitis flare events. No difference was observed in irHepatitis risk between HCC patients and non-HCC patients. ALT > 40 U/L was an independent risk factor for irHepatitis. Among 283 HBsAg-positive patients, six patients (2.1%) experienced HBVr. HCC patients had a higher risk of HBVr than non-HCC patients (4.4% vs. 0.6%). No specific risk factor for HBVr could be identified. However, none of the patients under nucleos/tide analogue (NUC) prophylaxis experienced HBVr in this study. CONCLUSIONS Under ICI treatment, HCC patients had a higher risk of hepatitis flare events than non-HCC patients. Abnormal baseline ALT levels are a risk factor for hepatic adverse events. NUC prophylaxis can minimise the risk of HBVr.
Collapse
Affiliation(s)
- Yi‐Ping Hung
- Department of OncologyTaipei Veterans General HospitalTaipeiTaiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung UniversityTaipeiTaiwan
- School of MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Pei‐Chang Lee
- School of MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
- Division of Gastroenterology and HepatologyTaipei Veterans General HospitalTaipeiTaiwan
| | - Yen‐Hwa Chang
- Department of UrologyTaipei Veterans General HospitalTaipeiTaiwan
| | - Muh‐Hwa Yang
- Department of OncologyTaipei Veterans General HospitalTaipeiTaiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung UniversityTaipeiTaiwan
- School of MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Chao‐Hua Chiu
- Department of Chest MedicineTaipei Veterans General HospitalTaipeiTaiwan
| | - Ming‐Huang Chen
- Department of OncologyTaipei Veterans General HospitalTaipeiTaiwan
- School of MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Keng‐Hsin Lan
- School of MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
- Division of Gastroenterology and HepatologyTaipei Veterans General HospitalTaipeiTaiwan
| | - I‐Cheng Lee
- School of MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
- Division of Gastroenterology and HepatologyTaipei Veterans General HospitalTaipeiTaiwan
| | - Ming‐Chih Hou
- School of MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Yee Chao
- Department of OncologyTaipei Veterans General HospitalTaipeiTaiwan
| | - Yi‐Hsiang Huang
- Institute of Clinical Medicine, National Yang Ming Chiao Tung UniversityTaipeiTaiwan
- School of MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
- Division of Gastroenterology and HepatologyTaipei Veterans General HospitalTaipeiTaiwan
- Healthcare and Services CenterTaipei Veterans General HospitalTaipeiTaiwan
| |
Collapse
|
135
|
Čerina Pavlinović D, Šuto Pavičić J, Njavro A, Librenjak N, Tomaš I, Šeparović R, Pleština S, Bajić Ž, Dedić Plavetić N, Vrdoljak E. Precision Oncology in Clinical Practice: Two Years of Comprehensive Genomic Profiling in Croatia. J Pers Med 2025; 15:59. [PMID: 39997336 PMCID: PMC11856208 DOI: 10.3390/jpm15020059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 01/17/2025] [Accepted: 01/30/2025] [Indexed: 02/26/2025] Open
Abstract
Background: The widespread adoption of precision medicine in routine cancer care remains a critical challenge, even as advanced technologies expand and personalized therapies demonstrate remarkable success in certain cancer types. While breakthrough innovations in targeted treatments have revolutionized outcomes for specific cancers, translating these scientific advances into standard clinical practice continues to be an evolving and complex endeavor. Croatia has a nationwide project of precision oncology through the comprehensive genomic profiling (CGP) analysis. Since collecting and analyzing real-world data is crucial for clinical research and defining the value of CGP in precision oncology, we aimed to present the data from everyday clinical practice given the opportunities and challenges we faced. Methods: This was a retrospective observational study conducted at the national level in all patients whose tumor samples were subjected to CGP between 1 January 2020 and 31 December 2021. Results: In total, 481 patients with CGP results were included in this study. Gastrointestinal and reproductive malignancies were the most common, accounting for 29.1% and 28.9% of all tested tumors, respectively. Specifically, colorectal tumors made up 19.1% of cases, while uterine tumors represented 11.2%. At least one clinically relevant genomic alteration was found in 76.7% of patients, with the KRAS mutation (27.2%) being the most common. During the two-year study period, 26,709 individuals lost their lives to cancer in Croatia. Combining this with the CGP selection criteria valid at the time, there was an estimated population of approximately 13,350 potentially eligible patients for the CGP analysis, meaning that only 3.6% of potentially eligible patients were tested. Conclusions: The analysis identified clinically actionable genomic alterations in approximately 80% of the evaluated patients, suggesting they could be candidates for targeted therapeutic interventions. The adoption of CGP remains limited, with estimates indicating that under 5% of metastatic cancer patients received testing in the initial two-year implementation period, despite established national insurance coverage guidelines. This low utilization rate suggests a significant gap in access to genomic testing, leaving many eligible cancer patients without the potential benefits of this diagnostic approach.
Collapse
Affiliation(s)
- Dora Čerina Pavlinović
- Department of Oncology, University Hospital Center Split, 21 000 Split, Croatia; (D.Č.P.); (J.Š.P.)
- School of Medicine, University of Split, 21 000 Split, Croatia
| | - Jelena Šuto Pavičić
- Department of Oncology, University Hospital Center Split, 21 000 Split, Croatia; (D.Č.P.); (J.Š.P.)
| | - Antonela Njavro
- Department of Oncology and Nuclear Medicine, Sestre Milosrdnice University Hospital Center, 10 000 Zagreb, Croatia;
| | - Nikša Librenjak
- Department of Oncology, University Hospital Center Zagreb, 10 000 Zagreb, Croatia; (N.L.); (S.P.)
| | - Ilijan Tomaš
- School of Medicine, Josip Juraj Strossmayer University of Osijek, 31 000 Osijek, Croatia;
| | - Robert Šeparović
- Department of Medical Oncology, University Hospital for Tumor, Sestre Milosrdnice, 10 000 Zagreb, Croatia;
| | - Stjepko Pleština
- Department of Oncology, University Hospital Center Zagreb, 10 000 Zagreb, Croatia; (N.L.); (S.P.)
- School of Medicine, University of Zagreb, 10 000 Zagreb, Croatia
| | - Žarko Bajić
- Research Unit “Dr. Mirko Grmek”, Psychiatric Clinic Sveti Ivan, 10 000 Zagreb, Croatia;
| | - Natalija Dedić Plavetić
- Department of Oncology, University Hospital Center Zagreb, 10 000 Zagreb, Croatia; (N.L.); (S.P.)
- School of Medicine, University of Zagreb, 10 000 Zagreb, Croatia
| | - Eduard Vrdoljak
- Department of Oncology, University Hospital Center Split, 21 000 Split, Croatia; (D.Č.P.); (J.Š.P.)
- School of Medicine, University of Split, 21 000 Split, Croatia
| |
Collapse
|
136
|
Strati A, Adamopoulos C, Kotsantis I, Psyrri A, Lianidou E, Papavassiliou AG. Targeting the PD-1/PD-L1 Signaling Pathway for Cancer Therapy: Focus on Biomarkers. Int J Mol Sci 2025; 26:1235. [PMID: 39941003 PMCID: PMC11818137 DOI: 10.3390/ijms26031235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 01/26/2025] [Accepted: 01/29/2025] [Indexed: 02/16/2025] Open
Abstract
The PD1/PD-L1 axis plays an important immunosuppressive role during the T-cell-mediated immune response, which is essential for the physiological homeostasis of the immune system. The biology of the immunological microenvironment is extremely complex and crucial for the development of treatment strategies for immunotherapy. Characterization of the immunological, genomic or transcriptomic landscape of cancer patients could allow discrimination between responders and non-responders to anti-PD-1/PD-L1 therapy. Immune checkpoint inhibitor (ICI) therapy has shown remarkable efficacy in a variety of malignancies in landmark trials and has fundamentally changed cancer therapy. Current research focuses on strategies to maximize patient selection for therapy, clarify mechanisms of resistance, improve existing biomarkers, including PD-L1 expression and tumor mutational burden (TMB), and discover new biomarkers. In this review, we focus on the function of the PD-1/PD-L1 signaling pathway and discuss the immunological, genomic, epigenetic and transcriptomic landscape in cancer patients receiving anti-PD-1/PD-L1 therapy. Finally, we provide an overview of the clinical trials testing the efficacy of antibodies against PD-1/PD-L1.
Collapse
Affiliation(s)
- Areti Strati
- Analysis of Circulating Tumor Cells, Lab of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece;
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (C.A.); (A.G.P.)
| | - Christos Adamopoulos
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (C.A.); (A.G.P.)
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ioannis Kotsantis
- Department of Medical Oncology, Second Department of Internal Medicine, Attikon University General Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Amanda Psyrri
- Department of Medical Oncology, Second Department of Internal Medicine, Attikon University General Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Evi Lianidou
- Analysis of Circulating Tumor Cells, Lab of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece;
| | - Athanasios G. Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (C.A.); (A.G.P.)
| |
Collapse
|
137
|
Nadelmann ER, Singh AK, Abbruzzese M, Adeuyan OO, Kenchappa DB, Kovrizhkin K, Lightman M, Samouha A, Tao KL, Yun J, Zhu TR, McLellan BN, Saenger YM. Acral Melanoma in Skin of Color: Current Insights and Future Directions: A Narrative Review. Cancers (Basel) 2025; 17:468. [PMID: 39941835 PMCID: PMC11816340 DOI: 10.3390/cancers17030468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/22/2025] [Accepted: 01/27/2025] [Indexed: 02/16/2025] Open
Abstract
Acral melanoma (AM), also known as acral lentiginous melanoma (ALM), is a rare subtype of melanoma that predominantly occurs on the palms, soles, and nail beds (Figure 1) [...].
Collapse
Affiliation(s)
- Emily R. Nadelmann
- Division of Dermatology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (E.R.N.); (M.A.); (K.K.); (M.L.); (A.S.); (K.L.T.); (J.Y.); (T.R.Z.); (B.N.M.)
| | - Ajay K. Singh
- Department of Oncology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (A.K.S.); (D.B.K.)
| | - Matteo Abbruzzese
- Division of Dermatology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (E.R.N.); (M.A.); (K.K.); (M.L.); (A.S.); (K.L.T.); (J.Y.); (T.R.Z.); (B.N.M.)
| | - Oluwaseyi O. Adeuyan
- Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA;
| | - Divya B. Kenchappa
- Department of Oncology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (A.K.S.); (D.B.K.)
| | - Katherine Kovrizhkin
- Division of Dermatology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (E.R.N.); (M.A.); (K.K.); (M.L.); (A.S.); (K.L.T.); (J.Y.); (T.R.Z.); (B.N.M.)
| | - Michelle Lightman
- Division of Dermatology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (E.R.N.); (M.A.); (K.K.); (M.L.); (A.S.); (K.L.T.); (J.Y.); (T.R.Z.); (B.N.M.)
| | - Avishai Samouha
- Division of Dermatology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (E.R.N.); (M.A.); (K.K.); (M.L.); (A.S.); (K.L.T.); (J.Y.); (T.R.Z.); (B.N.M.)
| | - Kevin L. Tao
- Division of Dermatology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (E.R.N.); (M.A.); (K.K.); (M.L.); (A.S.); (K.L.T.); (J.Y.); (T.R.Z.); (B.N.M.)
| | - Jaewon Yun
- Division of Dermatology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (E.R.N.); (M.A.); (K.K.); (M.L.); (A.S.); (K.L.T.); (J.Y.); (T.R.Z.); (B.N.M.)
| | - Tian R. Zhu
- Division of Dermatology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (E.R.N.); (M.A.); (K.K.); (M.L.); (A.S.); (K.L.T.); (J.Y.); (T.R.Z.); (B.N.M.)
| | - Beth N. McLellan
- Division of Dermatology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (E.R.N.); (M.A.); (K.K.); (M.L.); (A.S.); (K.L.T.); (J.Y.); (T.R.Z.); (B.N.M.)
| | - Yvonne M. Saenger
- Department of Oncology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (A.K.S.); (D.B.K.)
| |
Collapse
|
138
|
Hugdahl E, Aziz S, Klingen TA, Akslen LA. Prognostic value of immune biomarkers in melanoma loco-regional metastases. PLoS One 2025; 20:e0315284. [PMID: 39883679 PMCID: PMC11781691 DOI: 10.1371/journal.pone.0315284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 11/24/2024] [Indexed: 02/01/2025] Open
Abstract
The prognosis for patients with melanoma loco-regional metastases is very heterogenous. Adjuvant PD-L1-inhibitors have improved clinical outcome for this patient group, but the prognostic impact of tumour PD-L1 expression and number of tumour infiltrating lymphocytes (TILs) is still largely unknown. Here, we investigated the impact on survival for CD3, CD8, FOXP3 and PD-L1 TIL counts and tumour PD-L1 expression in melanoma loco-regional metastases. In a patient series of loco-regional metastases from nodular melanomas (n = 78; n = 26 skin metastases, n = 52 lymph node metastases), expression of PD-L1 in tumour cells and the number of CD3, CD8, FOXP3 and PD-L1 positive TILs were determined by immunohistochemistry on tissue microarray (TMA) slides. Due to limited tumour tissue in the paraffin blocks, 67 of the 78 cases were included for tissue microarrays. Low FOXP3 TIL count and negative tumour PD-L1 expression (cut off 1%) were both significantly associated with reduced survival in lymph node metastases. Low FOXP3 TIL count was significantly associated with low CD8, CD3 and PD-L1 TIL counts. Negative tumour PD-L1 expression was significantly associated with low CD8 and PD-L1 TIL count, large lymph node metastasis tumour size and presence of necrosis in lymph node metastases. Our findings demonstrate for the first time the negative prognostic value of low FOXP3 TIL count and confirm a negative prognostic value of negative tumour PD-L1 expression in melanoma lymph node metastases.
Collapse
Affiliation(s)
- Emilia Hugdahl
- Department of Clinical Medicine, Centre for Cancer Biomarkers CCBIO, University of Bergen, Bergen, Norway
| | - Sura Aziz
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Tor A. Klingen
- Department of Clinical Medicine, Centre for Cancer Biomarkers CCBIO, University of Bergen, Bergen, Norway
- Department of Pathology, Vestfold Hospital, Tønsberg, Norway
| | - Lars A. Akslen
- Department of Clinical Medicine, Centre for Cancer Biomarkers CCBIO, University of Bergen, Bergen, Norway
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
139
|
Greenwald NF, Nederlof I, Sowers C, Ding DY, Park S, Kong A, Houlahan KE, Varra SR, de Graaf M, Geurts V, Liu CC, Ranek JS, Voorwerk L, de Maaker M, Kagel A, McCaffrey E, Khan A, Yeh CY, Fullaway CC, Khair Z, Bai Y, Piyadasa H, Risom T, Delmastro A, Hartmann FJ, Mangiante L, Sotomayor-Vivas C, Schumacher TN, Ma Z, Bosse M, van de Vijver MJ, Tibshirani R, Horlings HM, Curtis C, Kok M, Angelo M. Temporal and spatial composition of the tumor microenvironment predicts response to immune checkpoint inhibition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.26.634557. [PMID: 39975273 PMCID: PMC11838242 DOI: 10.1101/2025.01.26.634557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Immune checkpoint inhibition (ICI) has fundamentally changed cancer treatment. However, only a minority of patients with metastatic triple negative breast cancer (TNBC) benefit from ICI, and the determinants of response remain largely unknown. To better understand the factors influencing patient outcome, we assembled a longitudinal cohort with tissue from multiple timepoints, including primary tumor, pre-treatment metastatic tumor, and on-treatment metastatic tumor from 117 patients treated with ICI (nivolumab) in the phase II TONIC trial. We used highly multiplexed imaging to quantify the subcellular localization of 37 proteins in each tumor. To extract meaningful information from the imaging data, we developed SpaceCat, a computational pipeline that quantifies features from imaging data such as cell density, cell diversity, spatial structure, and functional marker expression. We applied SpaceCat to 678 images from 294 tumors, generating more than 800 distinct features per tumor. Spatial features were more predictive of patient outcome, including features like the degree of mixing between cancer and immune cells, the diversity of the neighboring immune cells surrounding cancer cells, and the degree of T cell infiltration at the tumor border. Non-spatial features, including the ratio between T cell subsets and cancer cells and PD-L1 levels on myeloid cells, were also associated with patient outcome. Surprisingly, we did not identify robust predictors of response in the primary tumors. In contrast, the metastatic tumors had numerous features which predicted response. Some of these features, such as the cellular diversity at the tumor border, were shared across timepoints, but many of the features, such as T cell infiltration at the tumor border, were predictive of response at only a single timepoint. We trained multivariate models on all of the features in the dataset, finding that we could accurately predict patient outcome from the pre-treatment metastatic tumors, with improved performance using the on-treatment tumors. We validated our findings in matched bulk RNA-seq data, finding the most informative features from the on-treatment samples. Our study highlights the importance of profiling sequential tumor biopsies to understand the evolution of the tumor microenvironment, elucidating the temporal and spatial dynamics underlying patient responses and underscoring the need for further research on the prognostic role of metastatic tissue and its utility in stratifying patients for ICI.
Collapse
Affiliation(s)
- Noah F. Greenwald
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Cancer Biology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Iris Nederlof
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Cameron Sowers
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Daisy Yi Ding
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Seongyeol Park
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Alex Kong
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Kathleen E. Houlahan
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Manon de Graaf
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Veerle Geurts
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Candace C. Liu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jolene S. Ranek
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Leonie Voorwerk
- Division of Molecular Oncology & Immunology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Michiel de Maaker
- Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Adam Kagel
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Erin McCaffrey
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Aziz Khan
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Christine Yiwen Yeh
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Zumana Khair
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yunhao Bai
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Chemistry, Stanford University School of Humanities and Sciences, Stanford, CA, USA, Stanford University School of Humanities and Sciences, Stanford, CA, USA
| | - Hadeesha Piyadasa
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Tyler Risom
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Alea Delmastro
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Felix J. Hartmann
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- German Cancer Research Center (DKFZ), Heidelberg, Systems Immunology & Single-Cell Biology, Germany
| | - Lise Mangiante
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Ton N. Schumacher
- Division of Molecular Oncology & Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Zhicheng Ma
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Marc Bosse
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Robert Tibshirani
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
- Department of Statistics, Stanford University, Stanford, CA, USA
| | - Hugo M. Horlings
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Christina Curtis
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Marleen Kok
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Michael Angelo
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
140
|
Wiley N, Zecevic M, Ho V, Stolzberg MJ, Cox D, Soloff EV, Hall E, Wang CL. Dual-energy CT iodine concentration as a biomarker for immunotherapy treatment response in metastatic melanoma and renal cell carcinoma patients. Eur Radiol 2025:10.1007/s00330-025-11351-4. [PMID: 39873753 DOI: 10.1007/s00330-025-11351-4] [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: 08/09/2024] [Revised: 11/01/2024] [Accepted: 12/10/2024] [Indexed: 01/30/2025]
Abstract
OBJECTIVE To investigate the predictive value of tumor iodine concentration obtained with dual-energy CT (DECT) for treatment response in patients treated with immune checkpoint inhibitors (ICI). MATERIALS AND METHODS Retrospective single-center study of consecutive metastatic melanoma and renal cell carcinoma (RCC) patients undergoing first-line ICI treatment. The iodine concentration measurement time points include prior to initiation of therapy (baseline [BL]), after initiation (follow-up [FU1]), and either time point nearest to 12 months or at time of progression (final follow-up [FFU]). Target lesion DECT-based whole-volume tumor normalized iodine concentration average (NICave) and size measurements were obtained. Reference standard was individual lesion FFU status categorized as responders or nonresponders per RECIST 1.1. Logistic regression model assessed NICave change and FU1 lesion response as predictors of FFU lesion outcome. Model's performance was summarized with AUC. Intraclass correlation coefficient (ICC) summarized inter-rater agreement of NICave. RESULTS Forty-six patients were included (mean age 61 ± 11 years, 12 women; 16 melanoma). Sixty-four of 175 target lesions were confirmed nonresponders at FFU. In a multivariable model, lesion status at FU1 (odds ratio [OR]: 27.4, p < 0.001) and changes in NICave from BL to FU1 (OR: 2.42 per 1-SD increase, p = 0.019) were significant predictors of lesion status at FFU. The model's AUC was 0.86 (95% CI: 0.76-0.93). Inter-rater reliability of NICave was 0.98 (95% CI: 0.97-0.99). CONCLUSIONS Changes in iodine concentration from baseline to first follow-up improve identification of delayed responding metastatic melanoma and RCC lesions treated with immune checkpoint inhibitor, initially classified as nonresponders by size change. KEY POINTS Question How can pseudoprogression/delayed treatment response in metastatic renal cell carcinoma (RCC) and melanoma patients on first-line immune checkpoint inhibitors be accurately identified? Findings Combining iodine concentration change from Dual-energy CT (baseline to first follow-up) with RECIST-based lesion size change improved prediction of final lesion outcome. Clinical relevance DECT-based whole-volume tumor iodine concentration for target lesions is useful as a predictive imaging biomarker for distinguishing delayed response from true progression in patients with metastatic RCC and melanoma treated with first-line immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Natalie Wiley
- Department of Radiology, University of Washington, Seattle, WA, USA
| | - Mladen Zecevic
- Department of Radiology, University of Washington, Seattle, WA, USA
| | - Vivian Ho
- Department of Radiology, University of Washington, Seattle, WA, USA
| | | | - Danielle Cox
- Department of Radiology, University of Washington, Seattle, WA, USA
| | - Erik V Soloff
- Department of Radiology, University of Washington, Seattle, WA, USA
| | - Evan Hall
- Division of Hematology and Oncology, University of Washington, Seattle, WA, USA
| | - Carolyn L Wang
- Department of Radiology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
141
|
Santos Freitas JDA, da Silva Neto MM, Freitas de Lima CK, Amaral Boa Sorte NC, Bendicho MT, de Freitas Santos Júnior A. Safety profiles in the use of immune checkpoint inhibitors by patients with cancer and pre-existing autoimmune diseases. Med Clin (Barc) 2025; 164:53-60. [PMID: 39414554 DOI: 10.1016/j.medcli.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 10/18/2024]
Abstract
INTRODUCTION The treatment of cancer when associated with autoimmune diseases (AID) has been the subject of immunotherapy investigation, especially with the use of immune checkpoint inhibitors (ICI). Clinical studies have restricted the evaluation of its use in special populations such as patients with AID, leaving a gap regarding the safety of using immunotherapy. OBJECTIVE Discuss the safety of using ICI in patients with cancer and AID, in specialized oncology units, in the cities of Bahia, Brazil. METHODS Retrospective and quantitative cross-sectional study on immune-related adverse events (IRAE) to the use of ICI in patients with cancer and AID. RESULTS Patients (39 with cancer, and 14 with AID and cancer) were studied. Men (between 30 and 95 years old), melanoma and lung cancer and Hashimoto's thyroiditis were predominance. Pembrolizumab and Nivolumab (anti-PDL-1) were drugs most used. In general, patients using anti-PDL-1 with AID had IRAE with greater frequency and severity: Grade 1 (57%) and 3/4 grades (43%) reactions. The gastrointestinal system presented a greater IRAE in both groups, however in patients with AID more severe reactions were found (0% versus 60%). Patients with cancer and AID had higher rates of IRAE compared to patients without AID, respectively, of discontinuation (50% versus 18%) and interruption (85% versus 20%) of treatment. CONCLUSION IRAE increased in patients using ICI with cancer and AID. This suggests that the presence of IAD, in cancer patients, can increase the severity of IRAE. Therefore, the adoption of more appropriate therapeutic strategies is essential for better therapeutic results.
Collapse
Affiliation(s)
| | | | - Cleverton Kleiton Freitas de Lima
- Pharmaceutical Biotechnology Department, Faculty of Pharmacy, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | | |
Collapse
|
142
|
Hu Q, Xuan J, Wang L, Shen K, Gao Z, Zhou Y, Wei C, Gu J. Application of adoptive cell therapy in malignant melanoma. J Transl Med 2025; 23:102. [PMID: 39844295 PMCID: PMC11752767 DOI: 10.1186/s12967-025-06093-2] [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: 11/05/2024] [Accepted: 01/07/2025] [Indexed: 01/24/2025] Open
Abstract
Cutaneous melanoma is one of the most aggressive skin cancers originating from skin pigment cells. Patients with advanced melanoma suffer a poor prognosis and generally cannot benefit well from surgical resection and chemo/target therapy due to metastasis and drug resistance. Thus, adoptive cell therapy (ACT), employing immune cells with specific tumor-recognizing receptors, has emerged as a promising therapeutic approach to display on-tumor toxicity. This review discusses the application, efficacy, limitations, as well as future prospects of four commonly utilized approaches -including tumor-infiltrating lymphocytes, chimeric antigen receptor (CAR) T cell, engineered T-cell receptor T cells, and chimeric antigen receptor NK cells- in the context of malignant melanoma.
Collapse
Affiliation(s)
- Qianrong Hu
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
| | - Jiangying Xuan
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
| | - Lu Wang
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
| | - Kangjie Shen
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
| | - Zixu Gao
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
| | - Yuhong Zhou
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Chuanyuan Wei
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China.
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China.
| | - Jianying Gu
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China.
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
143
|
van Ravensteijn SG, Amir AL, Tauriello DVF, van Herpen CML, Boers-Sonderen MJ, Wesseling YJW, van Brussel AGC, Keizer DM, Verheul HMW, Bol KF. Exploring the relation between TGF-β pathway activity and response to checkpoint inhibition in patients with metastatic melanoma. Clin Exp Immunol 2025; 219:uxae108. [PMID: 39668127 PMCID: PMC11773812 DOI: 10.1093/cei/uxae108] [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: 06/25/2024] [Revised: 09/23/2024] [Accepted: 11/28/2024] [Indexed: 12/14/2024] Open
Abstract
INTRODUCTION Immune checkpoint inhibition (ICI) is highly effective for the treatment of melanoma, but intrinsic resistance is present in a subgroup of patients. TGF-β pathway activity may play a role in this resistance by preventing T-cells from entering the tumor microenvironment, causing immune escape. We investigated the association of TGF-β signal transduction pathway activity with resistance to ICI treatment in advanced melanoma. Furthermore, other pathway activities were analyzed to better understand their potential role in ICI resistance. METHOD The activity of 8 signaling pathways (TGF-β, Hedgehog, MAPK, AR, NOTCH, PI3K, JAK/STAT1-2, and NFkB) was analyzed from tumor tissue from patients with advanced melanoma. Pathway activity scores (PAS) were explored for associations with survival and response to ICI in 34 patients (19 non-responders and 15 responders). A second, independent method to investigate the predictive value of TGF-β pathway activation was conducted by determining levels of phosphorylated SMAD2. RESULTS The mean TGF-β PAS of responders vs non-responders was 53.9 vs 56.8 (P = 0.265). No significant relation with progression-free survival was detected for TGF-β activity (P = 0.078). No association between pSMAD2 staining and treatment response or survival was identified. In contrast, Hedgehog scores of responders versus non-responders were 35.7 vs 41.6 (P = 0.038). High Hedgehog PAS was the sole significant predictor of resistance to ICI (OR 0.88, P = 0.033) and worse progression-free survival (HR 1-1.1, P = 0.012). CONCLUSION TGF-β pathway activation showed no significant relation with treatment response to ICI or survival in patients with advanced melanoma. Hedgehog PAS was identified as a possible biomarker associated with both treatment response and survival.
Collapse
Affiliation(s)
| | - Avital L Amir
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Daniele V F Tauriello
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, the Netherlands
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, the Netherlands
| | - Carla M L van Herpen
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Marye J Boers-Sonderen
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | | | | | - Henk M W Verheul
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, the Netherlands
| | - Kalijn F Bol
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
144
|
Di Donato M, Cristiani CM, Capone M, Garofalo C, Madonna G, Passacatini LC, Ottaviano M, Ascierto PA, Auricchio F, Carbone E, Migliaccio A, Castoria G. Role of the androgen receptor in melanoma aggressiveness. Cell Death Dis 2025; 16:34. [PMID: 39837817 PMCID: PMC11751086 DOI: 10.1038/s41419-025-07350-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 12/18/2024] [Accepted: 01/14/2025] [Indexed: 01/23/2025]
Abstract
Malignant melanoma represents the fifth most common cancer in the world and its incidence is rising. Novel therapies targeting receptor tyrosine kinases, kinases and immune checkpoints have been employed with a significant improvement of the overall survival and long-term disease containment. Nevertheless, the disease often progresses and becomes resistant to the therapies. As such, the discovery of new targets and drugs for advanced melanoma still remains a difficult task. Gender disparities, with a female advantage in melanoma incidence and outcome, have been reported. Although emerging studies support the pro-tumorigenic role of androgen/androgen receptor axis in melanoma, the molecular bases of such evidence are still under intense investigation. We now report that ligand activation of the androgen receptor drives melanoma invasiveness and its escape from natural killer-mediated cytotoxic effect. By combining different experimental approaches, we observe that melanoma escape is mediated by the androgen-triggered shedding of the surface molecule MICA. Specific blockade of ADAM10 or androgen receptor impairs the androgen-induced MICA shedding and melanoma immune-escape. Further, the increase in MICA serum levels correlates with a poor outcome in melanoma patients treated with the anti-PD-1 monoclonal antibody, pembrolizumab. At last, melanoma cells depleted of the androgen receptor become more responsive to the most commonly used immunocheckpoint inhibitors, suggesting that the receptor dampens the immunotherapy efficacy. Taken together, our findings identify the androgen receptor as a diagnostic guidance in melanoma and support the repositioning of AR blockers in clinical management of patients.
Collapse
Affiliation(s)
- Marzia Di Donato
- Department of Precision Medicine, University of Campania 'L. Vanvitelli'- Via L. De Crecchio 7, 80138, Naples, Italy
| | - Costanza Maria Cristiani
- Neuroscience Research Center, Department of Medical and Surgical Sciences - 'Magna Graecia' University of Catanzaro, 88100, Catanzaro, Italy
| | - Mariaelena Capone
- Department of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS- Fondazione "G. Pascale", Napoli, Italy
| | - Cinzia Garofalo
- Department of Experimental and Clinical Medicine, 'Magna Graecia' University of Catanzaro, 88100, Catanzaro, Italy
| | - Gabriele Madonna
- Department of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS- Fondazione "G. Pascale", Napoli, Italy
| | | | - Margaret Ottaviano
- Department of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS- Fondazione "G. Pascale", Napoli, Italy
| | - Paolo Antonio Ascierto
- Department of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS- Fondazione "G. Pascale", Napoli, Italy
| | - Ferdinando Auricchio
- Department of Precision Medicine, University of Campania 'L. Vanvitelli'- Via L. De Crecchio 7, 80138, Naples, Italy
| | - Ennio Carbone
- Department of Precision Medicine, University of Campania 'L. Vanvitelli'- Via L. De Crecchio 7, 80138, Naples, Italy
| | - Antimo Migliaccio
- Department of Precision Medicine, University of Campania 'L. Vanvitelli'- Via L. De Crecchio 7, 80138, Naples, Italy.
| | - Gabriella Castoria
- Department of Precision Medicine, University of Campania 'L. Vanvitelli'- Via L. De Crecchio 7, 80138, Naples, Italy.
| |
Collapse
|
145
|
Buckley MW, Balaji Warner A, Brahmer J, Cappelli LC, Sharfman WH, Fuchs E, Kang H, Forde PM, Gladstone DE, Ambinder R, Kelly RJ, Lipson EJ, Gojo I, Lee EJ, Johnson TP, Saidha S, Llinas R, Ostrow LW, Naidoo J, Probasco JC. Immune-related encephalitis after immune checkpoint inhibitor therapy. Oncologist 2025; 30:oyae186. [PMID: 39066587 PMCID: PMC11783331 DOI: 10.1093/oncolo/oyae186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICI) have revolutionized cancer treatment but can trigger immune-related encephalitis. We report one of the largest case series of patients with immune-related encephalitis and review of the literature. METHODS Retrospective series of patients with immune-related encephalitis and literature review. RESULTS Fourteen patients with cancer treated with ICI (50% combination therapy) developed immune-related encephalitis. Diagnostic testing revealed cerebral spinal fluid (CSF) lymphocytic pleocytosis (85%) and elevated protein (69%), abnormal brain magnetic resonance imaging(MRI) (33%) or brain FDG-PET (25%), electroencephalogram (EEG) abnormalities (30%), and autoantibodies (31%). Encephalitis treatment included: corticosteroids (86%), intravenous immunoglobulin (IVIg) (36%), plasmapheresis (7%), and rituximab (29%). There were no deaths and 12 patients had significant recovery, although long-term complications were observed. All patients discontinued ICI. Longitudinal follow-up demonstrated anti-cancer response to ICI at 3 months (85%) and 6 months post-ICI initiation (77%). A literature review identified 132 patients with immune-related encephalitis. Most were treated with PD-1 inhibitors (18% combination). Common abnormalities included elevated CSF protein (84%) or pleocytosis (77%), abnormal brain MRI (65%), or autoantibodies (47%). Nearly all were treated with corticosteroids, many required additional therapy with IVIg (26%) or rituximab (12%). Most patients had clinical improvement (81%) but a minority (10%) had a clinical relapse after completing corticosteroid taper. ICIs were resumed in 7 patients (5%), with relapse in 3. CONCLUSIONS AND RELEVANCE Immune-related encephalitis is treatable and improves with corticosteroids in most cases but may require additional immunosuppression. Re-emergence of encephalitis is rare and does not typically result in adverse outcomes, and this should be considered in neurological immune-related adverse event management guidelines.
Collapse
Affiliation(s)
- Monica W Buckley
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
- Department of Neurology, University of Virginia School of Medicine, Charlottesville, VA 22903, United States
| | - Aanika Balaji Warner
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287, United States
| | - Julie Brahmer
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287, United States
| | - Laura C Cappelli
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - William H Sharfman
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287, United States
| | - Ephraim Fuchs
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Hyunseok Kang
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, United States
| | - Patrick M Forde
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287, United States
| | - Douglas E Gladstone
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
- R.J. Zuckerberg Cancer Center at Hofstra/Northwell Health, Lake Success, NY 11042, United States
| | - Richard Ambinder
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Ronan J Kelly
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
- Charles A. Sammons Cancer Center, Baylor University Medical Center, Dallas, TX 75246, United States
| | - Evan J Lipson
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287, United States
| | - Ivana Gojo
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Edward J Lee
- Maryland Oncology Hematology, Columbia, MD 21044, United States
| | - Tory P Johnson
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Shiv Saidha
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Rafael Llinas
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Lyle W Ostrow
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
- Department of Neurology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Jarushka Naidoo
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287, United States
- Department of Oncology, Johns Hopkins Bayview Medical Center, Baltimore, MD 21224, United States
- Department of Medicine, Beaumont Hospital Dublin and RCSI University of Health Sciences, Dublin, 9, Ireland
| | - John C Probasco
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| |
Collapse
|
146
|
Wang X, Ma S, Zhu S, Zhu L, Guo W. Advances in Immunotherapy and Targeted Therapy of Malignant Melanoma. Biomedicines 2025; 13:225. [PMID: 39857808 PMCID: PMC11761959 DOI: 10.3390/biomedicines13010225] [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: 12/02/2024] [Revised: 12/24/2024] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Malignant melanoma (MM) is a malignant tumor, resulting from mutations in melanocytes of the skin and mucous membranes. Its mortality rate accounts for 90% of all dermatologic tumor mortality. Traditional treatments such as surgery, chemotherapy, and radiotherapy are unable to achieve the expected results due to MM's low sensitivity, high drug resistance, and toxic side effects. As treatment advances, immunotherapy and targeted therapy have made significant breakthroughs in the treatment of MM and have demonstrated promising application prospects. However, the heterogeneity of tumor immune response causes more than half of patients to not benefit from clinical immunotherapy and targeted therapy, which delays the patient's condition and causes them to suffer adverse immune events' side effects. The combination of immunotherapy and targeted therapy can help improve therapeutic effects, delay drug resistance, and mitigate adverse effects. This review provides a comprehensive overview of the current development status and research progress of immune checkpoints, targeted genes, and their inhibitors, with a view to providing a reference for the clinical treatment of MM.
Collapse
Affiliation(s)
- Xue Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (X.W.); (S.M.); (S.Z.)
| | - Shanshan Ma
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (X.W.); (S.M.); (S.Z.)
| | - Shuting Zhu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (X.W.); (S.M.); (S.Z.)
| | - Liucun Zhu
- School of Life Sciences, Shanghai University, Shanghai 200444, China;
| | - Wenna Guo
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (X.W.); (S.M.); (S.Z.)
| |
Collapse
|
147
|
Liu L, Hou S, Zhu A, Yan B, Li L, Song D. The prognostic value of circulating tumor DNA in malignant melanoma patients treated with immune checkpoint inhibitors: a systematic review and meta-analysis. Front Immunol 2025; 15:1520441. [PMID: 39896816 PMCID: PMC11782251 DOI: 10.3389/fimmu.2024.1520441] [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: 11/06/2024] [Accepted: 12/30/2024] [Indexed: 02/04/2025] Open
Abstract
Background Circulating tumor DNA (ctDNA) is an emerging biomarker in malignant melanoma(MM), and high levels of ctDNA may reflect a higher tumor load. However, its prognostic value for MM receiving immune checkpoint inhibitors(ICI) remains controversial. This meta-analysis aimed to elucidate the prognostic significance of ctDNA in this patient population. Methods We conducted a comprehensive search of the PubMed, Cochrane Library, CNKI, and EMBASE databases, including studies published up to August 15, 2024, to investigate the prognostic impact of ctDNA in MM patients treated with ICI. Using a fixed-effects model, we systematically evaluated the association between ctDNA levels and key survival outcomes, including overall survival (OS) and progression-free survival (PFS). Additionally, funnel plots, Begg's test, and Egger's test were employed to assess potential publication bias. Results Twelve studies from eleven articles, involving a total of 1063 eligible MM patients receiving ICI therapy, were included. The results indicated that patients with detectable ctDNA before initiating ICI therapy had significantly poorer OS (HR = 3.19, 95% CI = 2.22-4.58, P < 0.001) and PFS (HR = 2.08, 95% CI = 1.61-2.69, P < 0.001). Furthermore, the detectability of ctDNA during treatment was also significantly associated with worse OS (HR = 4.57, 95% CI = 3.03-6.91, P < 0.001) and PFS (HR = 3.79, 95% CI = 2.13-6.75, P < 0.001). Conclusions This meta-analysis indicates that in MM patients receiving ICI therapy, detectable and high levels of ctDNA are significantly associated with poorer OS and PFS. Therefore, ctDNA can serve as a diagnostic and stratification tool prior to treatment, as well as an effective indicator for monitoring treatment response and disease progression. Systematic Review Registration www.inplasy.com, identifier INPLASY2024110018.
Collapse
Affiliation(s)
- Lei Liu
- Department of Neurology, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shufu Hou
- Department of Gastrointestinal Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Aiping Zhu
- Department of Neurology, Shandong Second Provincial General Hospital, Jinan, China
| | - Bing Yan
- Department of Gastrointestinal Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Linchuan Li
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Dandan Song
- Department of Neurology, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
148
|
Mok S, Liu H, Ağaç Çobanoğlu D, Anang NAAS, Mancuso JJ, Wherry EJ, Allison JP. Anti-CTLA-4 generates greater memory response than anti-PD-1 via TCF-1. Proc Natl Acad Sci U S A 2025; 122:e2418985122. [PMID: 39786926 PMCID: PMC11745370 DOI: 10.1073/pnas.2418985122] [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: 09/16/2024] [Accepted: 12/10/2024] [Indexed: 01/12/2025] Open
Abstract
The effects of T cell differentiation arising from immune checkpoint inhibition targeting cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and programmed cell death protein 1 (PD-1) on the immunological memory response remain unclear. Our investigation into the effects of anti-CTLA-4 and anti-PD-1 on memory T cell formation in mice reveals that memory T cells generated by anti-CTLA-4 exhibit greater expansion, cytokine production, and antitumor activity than those from anti-PD-1. Notably, anti-CTLA-4 preserves more T cell factor-1 (TCF-1)+ T cells during priming, while anti-PD-1 leads to more thymocyte selection-associated high mobility group box (TOX)+ T cells. Experiments using conditional Tcf7- or Tox-knockout mice highlight that TCF-1 is essential for the memory response generated by anti-CTLA-4, whereas TOX deletion alone in T cells has no effect on the response to anti-PD-1. Deepening our understanding of how checkpoint inhibition affects memory response is crucial for advancing our understanding of the enduring impacts of these immunotherapies on the immune system.
Collapse
Affiliation(s)
- Stephen Mok
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX77030
| | - Huey Liu
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX77030
| | - Didem Ağaç Çobanoğlu
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX77030
| | - Nana-Ama A. S. Anang
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX77030
| | - James J. Mancuso
- James P. Allison Institute, The University of Texas MD Anderson Cancer Center, Houston, TX77030
| | - E. John Wherry
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA19104
| | - James P. Allison
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX77030
- James P. Allison Institute, The University of Texas MD Anderson Cancer Center, Houston, TX77030
- Parker Institute for Cancer Immunotherapy, The University of Texas MD Anderson Cancer Center, Houston, TX77030
| |
Collapse
|
149
|
Manole S, Nguyen DH, Min JJ, Zhou S, Forbes N. Setting "cold" tumors on fire: Cancer therapy with live tumor-targeting bacteria. MED 2025; 6:100549. [PMID: 39689707 DOI: 10.1016/j.medj.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 09/18/2024] [Accepted: 11/01/2024] [Indexed: 12/19/2024]
Abstract
Immunotherapy with checkpoint blockade has shown remarkable efficacy in many patients with a variety of different types of cancer. However, the majority of patients with cancer have yet to benefit from this revolutionary therapy. Studies have shown that checkpoint blockade works best against immune-inflamed tumors characterized by the presence of tumor-infiltrating lymphocytes (TILs). In this review, we summarize studies using live tumor-targeting bacteria to treat cancer and describe various strategies to engineer the tumor-targeting bacteria for maximized immunoregulatory effects. We propose that tumor-localized infections by such engineered bacteria can create an immune microenvironment in favor of a more effective antitumor immunity with or without other therapies, such as immune checkpoint blockade (ICB). Finally, we will briefly outline some exemplary oncology clinical trials involving ICB plus live therapeutic bacteria, with a focus on their ability to modulate antitumor immune responses.
Collapse
Affiliation(s)
- Simin Manole
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA, USA
| | - Dinh-Huy Nguyen
- Institute for Molecular Imaging and Theranostics, Chonnam National University, Hwasun, Jeonnam 58128, South Korea
| | - Jung-Joon Min
- Institute for Molecular Imaging and Theranostics, Chonnam National University, Hwasun, Jeonnam 58128, South Korea; Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Jeonnam 58128, South Korea.
| | - Shibin Zhou
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287, USA; Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| | - Neil Forbes
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA, USA; Department of Chemical Engineering, University of Massachusetts, Amherst, MA, USA; Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, USA; Department of Microbiology, University of Massachusetts, Amherst, MA, USA.
| |
Collapse
|
150
|
Sun C, Cheng Y, Dong J, Hu L, Zhang Y, Shen H, Zhang G, Jiang B, Adam Youssouf S, Min W, Shen Y, Wang L, Deng H, Xiao Y, Yang P. Novel PD-L1/VISTA Dual Inhibitor as Potential Immunotherapy Agents. J Med Chem 2025; 68:156-173. [PMID: 39731560 DOI: 10.1021/acs.jmedchem.4c01640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2024]
Abstract
Inhibiting the activity of immune checkpoint proteins to reignite the antitumor activity of immune cells has emerged as a pivotal strategy. PD-L1 and VISTA, as critical proteins governing immune regulation, are concurrently upregulated under conditions such as hypoxia. Through a rational drug design process, P17, a dual-target inhibitor for PD-L1 and VISTA is identified. This inhibitor blocks the signaling pathways of both PD-L1 and VISTA at the protein and cellular levels, thereby reactivating the antitumor function of T cells. P17 displays encouraging attributes in terms of druggability and safety assessments. Notably, P17 demonstrates superior antitumor efficacy compared to single-target inhibitors at equivalent doses in in vivo experiments. More crucially, P17 significantly enhances the infiltration of immune cells. This study not only validates the effectiveness of a dual-target inhibitor strategy against PD-L1 and VISTA, but also identifies P17 as a promising candidate molecule with significant therapeutic potential.
Collapse
Affiliation(s)
- Chengliang Sun
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Yao Cheng
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Jingwen Dong
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Lingrong Hu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Yu Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR 999077, China
| | - Hao Shen
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Guoyu Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Binjian Jiang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Salouoi Adam Youssouf
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Wenjian Min
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Yuxia Shen
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Liping Wang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Hongbin Deng
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yibei Xiao
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Chongqing Innovation Institute of China Pharmaceutical University, Chongqing 401135, China
| | - Peng Yang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|