1
|
Zheng X, Sun R, Wei T. Immune microenvironment in papillary thyroid carcinoma: roles of immune cells and checkpoints in disease progression and therapeutic implications. Front Immunol 2024; 15:1438235. [PMID: 39290709 PMCID: PMC11405226 DOI: 10.3389/fimmu.2024.1438235] [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: 05/25/2024] [Accepted: 08/14/2024] [Indexed: 09/19/2024] Open
Abstract
Papillary thyroid cancer (PTC) is the most common type of primary thyroid cancer. Despite the low malignancy and relatively good prognosis, some PTC cases are highly aggressive and even develop refractory cancer in the thyroid. Growing evidence suggested that microenvironment in tumor affected PTC biological behavior due to different immune states. Different interconnected components in the immune system influence and participate in tumor invasion, and are closely related to PTC metastasis. Immune cells and molecules are widely distributed in PTC tissues. Their quantity and proportion vary with the host's immune status, which suggests that immunotherapy may be a very promising therapeutic modality for PTC. In this paper, we review the role of immune cells and immune checkpoints in PTC immune microenvironment based on the characteristics of the PTC tumor microenvironment.
Collapse
Affiliation(s)
- Xun Zheng
- Department of Thyroid and Parathyroid Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Ruonan Sun
- Department of Thyroid and Parathyroid Surgery, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Tao Wei
- Department of Thyroid and Parathyroid Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
2
|
Li S, Sheng J, Zhang D, Qin H. Targeting tumor-associated macrophages to reverse antitumor drug resistance. Aging (Albany NY) 2024; 16:10165-10196. [PMID: 38787372 PMCID: PMC11210230 DOI: 10.18632/aging.205858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 04/22/2024] [Indexed: 05/25/2024]
Abstract
Currently, antitumor drugs show limited clinical outcomes, mainly due to adaptive resistance. Clinical evidence has highlighted the importance of the tumor microenvironment (TME) and tumor-associated macrophages (TAMs) in tumor response to conventional antitumor drugs. Preclinical studies show that TAMs following antitumor agent can be reprogrammed to an immunosuppressive phenotype and proangiogenic activities through different mechanisms, mediating drug resistance and poor prognosis. Potential extrinsic inhibitors targeting TAMs repolarize to an M1-like phenotype or downregulate proangiogenic function, enhancing therapeutic efficacy of anti-tumor therapy. Moreover, pharmacological modulation of macrophages that restore the immune stimulatory characteristics is useful to reshaping the tumor microenvironment, thus further limiting tumor growth. This review aims to introduce macrophage response in tumor therapy and provide a potential therapeutic combination strategy of TAM-targeting immunomodulation with conventional antitumor drugs.
Collapse
Affiliation(s)
- Sheng Li
- The Second Hospital of Jilin University, Changchun, China
| | - Jiyao Sheng
- Department of Hepatobiliary and Pancreatic Surgery, Second Hospital of Jilin University, Changchun, China
| | - Dan Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Second Hospital of Jilin University, Changchun, China
| | - Hanjiao Qin
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
3
|
Ducoulombier A, Guigay J, Etienne-Grimaldi MC, Saada-Bouzid E. Chemotherapy postimmunotherapy for recurrent metastatic head and neck squamous cell carcinoma. Curr Opin Oncol 2023; 35:166-177. [PMID: 36966495 DOI: 10.1097/cco.0000000000000936] [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: 03/27/2023]
Abstract
PURPOSE OF REVIEW Clinical data on salvage chemotherapy used after checkpoints inhibitors in oncology are reviewed, with a special focus on recurrent/metastatic head and neck squamous cell carcinoma (R/M HNSCC). RECENT FINDINGS Converging evidence is emerging about high response and/or disease control rates associated with salvage chemotherapy after immunotherapy failure in advanced solid tumours. This phenomenon is mainly reported in retrospective studies for "hot tumours" such as R/M HNSCC, melanoma, lung, urothelial or gastric cancers, but also in haematological malignancies. Some physiopathological hypotheses have been raised. SUMMARY Several independent series show increased response rates associated with postimmuno chemotherapy when compared with retrospective series in similar settings. Several mechanisms could be involved such as a "carry-over" allowed by a persistence of the checkpoint inhibitor, a modulation of tumour microenvironment components but also an intrinsic immunomodulatory effect of chemotherapy, increased by a specific immunologic state induced by the therapeutic pressure of checkpoint inhibitors. These data establish a rationale for prospectively evaluating the features of postimmunotherapy salvage chemotherapy.
Collapse
Affiliation(s)
- Agnes Ducoulombier
- Laboratoire de Recherche Translationnelle en Oncologie, Université Côte d'Azur, Centre Antoine Lacassagne
- Department of Medical Oncology, Université Côte d'Azur, Centre Antoine Lacassagne, Nice, France
| | - Joel Guigay
- Department of Medical Oncology, Université Côte d'Azur, Centre Antoine Lacassagne, Nice, France
| | | | - Esma Saada-Bouzid
- Laboratoire de Recherche Translationnelle en Oncologie, Université Côte d'Azur, Centre Antoine Lacassagne
- Department of Medical Oncology, Université Côte d'Azur, Centre Antoine Lacassagne, Nice, France
| |
Collapse
|
4
|
Uchimoto T, Fukushima T, Komura K, Fukuokaya W, Adachi T, Hashimoto T, Yoshizawa A, Nakamura K, Yano Y, Nishimura K, Nishio K, Nakamori K, Iwatani K, Yamamoto S, Urabe F, Mori K, Yanagisawa T, Tsuduki S, Takahara K, Inamoto T, Miki J, Kimura T, Ohno Y, Shiroki R, Egawa S, Azuma H. Re-challenging chemotherapy after pembrolizumab in platinum-refractory urothelial carcinoma. BJU Int 2023; 131:477-485. [PMID: 36098556 DOI: 10.1111/bju.15893] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
OBJECTIVES To assess the real-world clinical benefit of re-challenging chemotherapy after pembrolizumab in patients with metastatic urothelial carcinoma (mUC), as there have been several reports suggesting that programmed cell death protein-1/programmed death-ligand 1inhibitors can restore platinum sensitivity. PATIENTS AND METHODS Of 236 patients treated with pembrolizumab, we excluded 45 patients who did not experience progressive disease (PD) for pembrolizumab during the follow-up and 86 patients who discontinued pembrolizumab by the diagnosis of PD followed by the best supportive care. A total of 105 patients were identified for a logistic regression propensity score model to compare the survival outcomes between patients treated with continuing pembrolizumab (80) and re-challenging chemotherapy (25) after the diagnosis of PD for pembrolizumab. RESULTS A median overall survival (OS) from PD for pembrolizumab was 11 months in 105 patients. Of 25 patients treated with re-challenging chemotherapy, platinum-including chemotherapy (gemcitabine and cisplatin; gemcitabine/cisplatin/paclitaxel [GCP]; methotrexate and vinblastine and adriamycin and cisplatin; and methotrexate and carboplatin and vinblastine MCAVI) was offered in 20 patients (80%). The objective response rate (ORR) for the first-line chemotherapy in the 105 patients was 30%, with a comparable ORR in 25 patients treated with re-challenging chemotherapy of 28%. GCP as a re-challenging regimen was offered in 12 of 25 (48%) patients. The ORR for the GCP regimen was 50%. Propensity score matching was performed using putative clinical factors, from which 34 patients were identified as pair-matched groups. The OS for patients treated with re-challenging chemotherapy was significantly longer than continuing pembrolizumab (a median of 13.9 and 5.8 months, respectively: P = 0.048). CONCLUSION Re-challenging chemotherapy including platinum agents after PD with pembrolizumab offers clinical benefits in patients with mUC.
Collapse
Affiliation(s)
- Taizo Uchimoto
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan
| | - Tatsuo Fukushima
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan
| | - Kazumasa Komura
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan
| | - Wataru Fukuokaya
- Department of Urology, The Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Takahiro Adachi
- Department of Urology, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Takeshi Hashimoto
- Department of Urology, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Atsuhiko Yoshizawa
- Department of Urology, Fujita-Health University School of Medicine, Toyoake City, Aichi, Japan
| | - Ko Nakamura
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan
| | - Yusuke Yano
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan
| | - Kazuki Nishimura
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan
| | - Kyosuke Nishio
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan
| | - Keita Nakamori
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan
| | - Kosuke Iwatani
- Department of Urology, The Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Shutaro Yamamoto
- Department of Urology, The Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Fumihiko Urabe
- Department of Urology, The Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Keiichiro Mori
- Department of Urology, The Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Takafumi Yanagisawa
- Department of Urology, The Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Shunsuke Tsuduki
- Department of Urology, The Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Kiyoshi Takahara
- Department of Urology, Fujita-Health University School of Medicine, Toyoake City, Aichi, Japan
| | - Teruo Inamoto
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan
| | - Jun Miki
- Department of Urology, The Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Takahiro Kimura
- Department of Urology, The Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Yoshio Ohno
- Department of Urology, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Ryoichi Shiroki
- Department of Urology, Fujita-Health University School of Medicine, Toyoake City, Aichi, Japan
| | - Shin Egawa
- Department of Urology, The Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Haruhito Azuma
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan
| |
Collapse
|
5
|
Meynard L, Dinart D, Delaunay B, Fléchon A, Saldana C, Lefort F, Gravis G, Thiery-Vuillemin A, Cancel M, Coquan E, Ladoire S, Maillet D, Rolland F, Boughalem E, Martin S, Laramas M, Crouzet L, Abbar B, Falkowski S, Pouessel D, Roubaud G. Chemotherapy following immune checkpoint inhibitors in patients with locally advanced or metastatic urothelial carcinoma. Eur J Cancer 2022; 175:43-53. [PMID: 36088671 DOI: 10.1016/j.ejca.2022.08.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 08/07/2022] [Accepted: 08/11/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Recent studies suggest improvements in response to salvage chemotherapy (CT) after immune checkpoint inhibitors (ICIs) in several types of cancer. Our objective was to assess the efficacy of chemotherapy re-challenge after ICI, compared with second-line chemotherapy without previous ICI in patients with locally advanced or metastatic urothelial carcinoma (la/mUC). METHODS In this multicentre retrospective study, we included all patients with la/mUC initiating second or third-line chemotherapy from January 2015 to June 2020. We compared patients treated with second-line chemotherapy without previous ICI (CT2) and patients treated with third-line chemotherapy after ICI (CT3). The primary end-point was objective response rate (ORR) in CT3 compared with CT2. Secondary end-points included progression-free survival (PFS) and toxicities. RESULTS Overall, 553 patients were included. ORRs were 31.0% (95% CI, 26.5 to 35.5) and 29.2% (95% CI, 21.9 to 36.6), respectively, in CT2 and CT3, with no statistically significant differences (P = 0.62). In subgroup analyses, no differences in ORR were observed by Bellmunt risk group, type of chemotherapy (platinum or taxanes), duration of response to first-platinum-based chemotherapy (< or ≥ 12 months) or FGFR-status. Median PFS was 4.6 months (95% CI, 3.9 to 5.1) and 4.9 months (95% CI, 4.1 to 5.5) in CT2 and CT3, respectively, and grade 3-4 hematologic toxicity occurred in 35.0% and 22.4% of patients. CONCLUSION This large multicentre retrospective study provides clinically relevant real-world data. Chemotherapy re-challenge after ICI in la/mUC achieves ORR and PFS comparable with those obtained in CT2 with an acceptable safety profile. These updated results offer more promising outcomes than historically reported with second-line chemotherapy data.
Collapse
Affiliation(s)
- Lucie Meynard
- Department of Medical Oncology, Institut Bergonié, Bordeaux, France.
| | - Derek Dinart
- University Bordeaux, Inserm, Bordeaux Population Health Research Center, Epicene Team, UMR 1219, F-33000 Bordeaux, France; Inserm CIC1401, Clinical and Epidemiological Research Unit, Institut Bergonié, Comprehensive Cancer Center, F-33000 Bordeaux, France
| | - Blandine Delaunay
- Department of Medical Oncology, Institut Claudius Régaud, IUCT Oncopole, Toulouse, France
| | - Aude Fléchon
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France
| | - Carolina Saldana
- AP-HP, Hopital Henri Mondor, Service d'Oncologie, Univ Paris Est Creteil, TRePCa, F-94010 Creteil, France
| | - Félix Lefort
- Department of Medical Oncology, University Hospital, Bordeaux, France
| | - Gwenaëlle Gravis
- Department of Medical Oncology, Institut Paoli Calmettes, Marseille, France
| | | | - Mathilde Cancel
- Department of Medical Oncology, University Hospital, Tours, France
| | - Elodie Coquan
- Department of Medical Oncology, Centre François Baclesse, Caen, France
| | - Sylvain Ladoire
- Department of Medical Oncology, Centre Georges François Leclerc, Dijon, France
| | - Denis Maillet
- Department of Medical Oncology, Institut de Cancérologie des Hospices Civils de Lyon (IC-HCL), Lyon, France
| | - Frédéric Rolland
- Department of Medical Oncology, Institut de Cancérologie de l'Ouest, Nantes, France
| | - Elouen Boughalem
- Department of Medical Oncology, Institut de Cancérologie de l'Ouest, Angers, France
| | - Sophie Martin
- Department of Medical Oncology Institut de Cancérologie Strasbourg Europe, Strasbourg, France
| | - Mathieu Laramas
- Department of Medical Oncology, University Hospital, Grenoble, France
| | - Laurence Crouzet
- Department of Medical Oncology, Centre Eugène Marquis, Rennes, France
| | - Baptiste Abbar
- Sorbonne Université, INSERM, Institut Pierre Louis d'Epidémiologie et de Santé Publique (iPLESP), Assistance Publique-Hôpitaux de Paris (AP-HP), Pitié Salpêtrière Hospital, Department of Medical Oncology, Institute Universitaire de Cancérologie, CLIP(2) Galilée, Paris, France
| | - Sabrina Falkowski
- Department of Medical Oncology, Clinique François Chénieux, Limoges, France
| | - Damien Pouessel
- Department of Medical Oncology, Institut Claudius Régaud, IUCT Oncopole, Toulouse, France
| | - Guilhem Roubaud
- Department of Medical Oncology, Institut Bergonié, Bordeaux, France
| |
Collapse
|
6
|
Baci D, Cekani E, Imperatori A, Ribatti D, Mortara L. Host-Related Factors as Targetable Drivers of Immunotherapy Response in Non-Small Cell Lung Cancer Patients. Front Immunol 2022; 13:914890. [PMID: 35874749 PMCID: PMC9298844 DOI: 10.3389/fimmu.2022.914890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 05/13/2022] [Indexed: 11/13/2022] Open
Abstract
Despite some significant therapeutic breakthroughs leading to immunotherapy, a high percentage of patients with non-small cell lung cancer (NSCLC) do not respond to treatment on relapse, thus experiencing poor prognosis and survival. The unsatisfying results could be related to the features of the tumor immune microenvironment and the dynamic interactions between a tumor and immune infiltrate. Host-tumor interactions strongly influence the course of disease and response to therapies. Thus, targeting host-associated factors by restoring their physiologic functions altered by the presence of a tumor represents a new therapeutic approach to control tumor development and progression. In NSCLC, the immunogenic tumor balance is shifted negatively toward immunosuppression due to the release of inhibitory factors as well as the presence of immunosuppressive cells. Among these cells, there are myeloid-derived suppressor cells, regulatory T cells that can generate a tumor-permissive milieu by reprogramming the cells of the hosts such as tumor-associated macrophages, tumor-associated neutrophils, natural killer cells, dendritic cells, and mast cells that acquire tumor-supporting phenotypes and functions. This review highlights the current knowledge of the involvement of host-related factors, including innate and adaptive immunity in orchestrating the tumor cell fate and the primary resistance mechanisms to immunotherapy in NSCLC. Finally, we discuss combinational therapeutic strategies targeting different aspects of the tumor immune microenvironment (TIME) to prime the host response. Further research dissecting the characteristics and dynamic interactions within the interface host-tumor is necessary to improve a patient fitness immune response and provide answers regarding the immunotherapy efficacy, with the aim to develop more successful treatments for NSCLC.
Collapse
Affiliation(s)
- Denisa Baci
- Molecular Cardiology Laboratory, IRCCS-Policlinico San Donato, San Donato Milanese, Milan, Italy.,Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Elona Cekani
- Medical Oncology Clinic, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Andrea Imperatori
- Center for Thoracic Surgery, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Aldo Moro Medical School, Bari, Italy
| | - Lorenzo Mortara
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| |
Collapse
|
7
|
Kwiatkowska I, Hermanowicz JM, Iwinska Z, Kowalczuk K, Iwanowska J, Pawlak D. Zebrafish—An Optimal Model in Experimental Oncology. Molecules 2022; 27:molecules27134223. [PMID: 35807468 PMCID: PMC9268704 DOI: 10.3390/molecules27134223] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/10/2022] [Accepted: 06/28/2022] [Indexed: 02/02/2023] Open
Abstract
A thorough understanding of cancer pathogenesis is a necessary step in the development of more effective and safer therapy. However, due to the complexity of the process and intricate interactions, studying tumor development is an extremely difficult and challenging task. In bringing this issue closer, different scientific models with various advancement levels are helpful. Cell cultures is a system that is too simple and does not allow for multidirectional research. On the other hand, rodent models, although commonly used, are burdened with several limitations. For this reason, new model organisms that will allow for the studying of carcinogenesis stages and factors reliably involved in them are urgently sought after. Danio rerio, an inconspicuous fish endowed with unique features, is gaining in importance in the world of scientific research. Including it in oncological research brings solutions to many challenges afflicting modern medicine. This article aims to illustrate the usefulness of Danio rerio as a model organism which turns out to be a powerful and unique tool for studying the stages of carcinogenesis and solving the hitherto incomprehensible processes that lead to the development of the disease.
Collapse
Affiliation(s)
- Iwona Kwiatkowska
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2C, 15-222 Bialystok, Poland; (J.M.H.); (Z.I.); (J.I.); (D.P.)
- Correspondence: ; Tel./Fax: +48-8574-856-01
| | - Justyna Magdalena Hermanowicz
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2C, 15-222 Bialystok, Poland; (J.M.H.); (Z.I.); (J.I.); (D.P.)
- Department of Clinical Pharmacy, Medical University of Bialystok, Mickiewicza 2C, 15-222 Bialystok, Poland
| | - Zaneta Iwinska
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2C, 15-222 Bialystok, Poland; (J.M.H.); (Z.I.); (J.I.); (D.P.)
| | - Krystyna Kowalczuk
- Department of Integrated Medical Care, Medical University of Bialystok, ul. M Skłodowskiej-Curie 7A, 15-096 Bialystok, Poland;
| | - Jolanta Iwanowska
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2C, 15-222 Bialystok, Poland; (J.M.H.); (Z.I.); (J.I.); (D.P.)
| | - Dariusz Pawlak
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2C, 15-222 Bialystok, Poland; (J.M.H.); (Z.I.); (J.I.); (D.P.)
| |
Collapse
|
8
|
Normalization of tumor vasculature: A potential strategy to increase the efficiency of immune checkpoint blockades in cancers. Int Immunopharmacol 2022; 110:108968. [PMID: 35764018 DOI: 10.1016/j.intimp.2022.108968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 11/23/2022]
Abstract
Immune checkpoint inhibitors (ICIs) eliminate tumor cells by reactivating CD8 + T cells using the cytotoxic effects of the immune system. However, in this process, tumor angiogenic factors and abnormal formation of tumor blood vessels are not conducive to the treatment of ICIs. In the tumor microenvironment (TME), proangiogenic factors prevent dendritic cell maturation, reduce T cell infiltration, and recruit inhibitory immune cells such as regulatory T (Treg) cells. Abnormal tumor blood vessels also prevent immune cells and chemotherapy drugs from reaching the target effectively and lead to poor perfusion and severe hypoxia of the tumor. Treatment with antiangiogenic inhibitors can block the transmission of abnormal angiogenesis signals and promote the normalization of tumor vasculature. Therefore, the combination of antiangiogenic inhibitors and ICIs is used in clinical therapy. Combination therapy has been proven theoretically feasible in preclinical trials, and many clinical trials have been completed to confirm its safety and efficacy.
Collapse
|
9
|
Hu H, Chen Y, Tan S, Wu S, Huang Y, Fu S, Luo F, He J. The Research Progress of Antiangiogenic Therapy, Immune Therapy and Tumor Microenvironment. Front Immunol 2022; 13:802846. [PMID: 35281003 PMCID: PMC8905241 DOI: 10.3389/fimmu.2022.802846] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 02/01/2022] [Indexed: 02/05/2023] Open
Abstract
Anti-angiogenesis therapy, a promising strategy against cancer progression, is limited by drug-resistance, which could be attributed to changes within the tumor microenvironment. Studies have increasingly shown that combining anti-angiogenesis drugs with immunotherapy synergistically inhibits tumor growth and progression. Combination of anti-angiogenesis therapy and immunotherapy are well-established therapeutic options among solid tumors, such as non-small cell lung cancer, hepatic cell carcinoma, and renal cell carcinoma. However, this combination has achieved an unsatisfactory effect among some tumors, such as breast cancer, glioblastoma, and pancreatic ductal adenocarcinoma. Therefore, resistance to anti-angiogenesis agents, as well as a lack of biomarkers, remains a challenge. In this review, the current anti-angiogenesis therapies and corresponding drug-resistance, the relationship between tumor microenvironment and immunotherapy, and the latest progress on the combination of both therapeutic modalities are discussed. The aim of this review is to discuss whether the combination of anti-angiogenesis therapy and immunotherapy can exert synergistic antitumor effects, which can provide a basis to exploring new targets and developing more advanced strategies.
Collapse
Affiliation(s)
- Haoyue Hu
- Lung Cancer Center, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, China.,Department of Medical Oncology, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Medicine School of University of Electronic Science and Technology, Chengdu, China
| | - Yue Chen
- Department of Pathology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Songtao Tan
- Lung Cancer Center, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, China
| | - Silin Wu
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yan Huang
- Lung Cancer Center, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, China
| | - Shengya Fu
- Lung Cancer Center, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, China.,Second Department of Oncology, Sichuan Friendship Hospital, Chengdu, China
| | - Feng Luo
- Lung Cancer Center, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, China
| | - Jun He
- Department of Oncology, The Third Hospital of Mianyang (Sichuan Mental Health Center), Mianyang, China
| |
Collapse
|
10
|
Sunaga N, Miura Y, Kasahara N, Sakurai R. Targeting Oncogenic KRAS in Non-Small-Cell Lung Cancer. Cancers (Basel) 2021; 13:cancers13235956. [PMID: 34885068 PMCID: PMC8656763 DOI: 10.3390/cancers13235956] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 11/24/2021] [Accepted: 11/24/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary v-Ki-ras2 Kirsten rat sarcoma viral oncogene (KRAS) is the most common driver in NSCLC, and targeting oncogenic KRAS is a major challenge in the treatment of non-small-cell lung cancer (NSCLC). While several covalent KRAS G12C inhibitors have emerged as a novel anti-KRAS therapy, the development of combined therapies involving the targeting of oncogenic KRAS plus other targeted drugs is still required given the vast heterogeneity of KRAS-mutated tumors. In this review, we summarize the biological and immunological characteristics of oncogenic KRAS-driven NSCLC and the preclinical and clinical evidence for mutant KRAS-targeted therapies. We also discuss the mechanisms of resistance to KRAS G12C inhibitors and possible therapeutic strategies to overcome this drug resistance. Abstract Recent advances in molecular biology and the resultant identification of driver oncogenes have achieved major progress in precision medicine for non-small-cell lung cancer (NSCLC). v-Ki-ras2 Kirsten rat sarcoma viral oncogene (KRAS) is the most common driver in NSCLC, and targeting KRAS is considerably important. The recent discovery of covalent KRAS G12C inhibitors offers hope for improving the prognosis of NSCLC patients, but the development of combination therapies corresponding to tumor characteristics is still required given the vast heterogeneity of KRAS-mutated NSCLC. In this review, we summarize the current understanding of KRAS mutations regarding the involvement of malignant transformation and describe the preclinical and clinical evidence for targeting KRAS-mutated NSCLC. We also discuss the mechanisms of resistance to KRAS G12C inhibitors and possible combination treatment strategies to overcome this drug resistance.
Collapse
Affiliation(s)
- Noriaki Sunaga
- Department of Respiratory Medicine, Gunma University Graduate School of Medicine, 3-39-15 Showa-machi, Maebashi 371-8511, Gunma, Japan;
- Correspondence: ; Tel.: +81-27-220-8000
| | - Yosuke Miura
- Department of Respiratory Medicine, Gunma University Graduate School of Medicine, 3-39-15 Showa-machi, Maebashi 371-8511, Gunma, Japan;
| | - Norimitsu Kasahara
- Innovative Medical Research Center, Gunma University Hospital, 3-39-15 Showa-machi, Maebashi 371-8511, Gunma, Japan;
| | - Reiko Sakurai
- Oncology Center, Gunma University Hospital, 3-39-15 Showa-machi, Maebashi 371-8511, Gunma, Japan;
| |
Collapse
|
11
|
Qureshi A, Michel M, Lerner J, Dasanu CA. Evolving therapeutic strategies for advanced hepatocellular carcinoma. Expert Opin Pharmacother 2021; 22:2495-2506. [PMID: 34252328 DOI: 10.1080/14656566.2021.1953473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Introduction: While sorafenib dominated the therapeutic arena in advanced hepatocellular carcinoma (HCC) for almost a decade, newer agents and combinations have been changing the therapeutic landscape in the last years.Areas covered: The authors outline the etiopathogenesis and evaluate the diagnostics in HCC, followed by a comprehensive review of the currently approved and experimental treatment approaches, with a focus on various systemic agents and combinations of agents. The manuscript was subdivided into relevant categories, thus making it applicable for both clinical practice and research endeavors.Expert opinion: Recently, combination therapies including immune checkpoint inhibitors with anti-VEGF/R agents have shown superior clinical efficacy in HCC. The Atezolizumab-bevacizumab combination is currently the preferred first-line therapy. Single-agents cabozantinib and regorafenib as well as nivolumab-ipilimumab combination are favored as second-line therapies. Further research is needed to identify the predictors of response to various treatments and establish the distinct patient profiles that will derive most benefit. Tumor mutation analysis and germline mutation testing could identify rational therapeutic targets in HCC in the near future. As the skyline for new therapeutic agents and combinations in HCC continues to expand, the outlook as of today is cautiously optimistic in this still difficult-to-treat malignant neoplastic disease.
Collapse
Affiliation(s)
- Ammar Qureshi
- Department of Internal Medicine, Eisenhower Health, Rancho Mirage, CA, USA
| | - Miguel Michel
- Department of Internal Medicine, Eisenhower Health, Rancho Mirage, CA, USA
| | - Jaren Lerner
- Department of Internal Medicine, Eisenhower Health, Rancho Mirage, CA, USA
| | - Constantin A Dasanu
- Department of Hematology-Oncology, Eisenhower Lucy Curci Cancer Center, Rancho Mirage, CA, USA.,Department of Hematology-Oncology, UC San Diego Health, San Diego, CA, USA
| |
Collapse
|
12
|
Liu X, Lu Y, Qin S. Atezolizumab and bevacizumab for hepatocellular carcinoma: mechanism, pharmacokinetics and future treatment strategies. Future Oncol 2021; 17:2243-2256. [PMID: 33663220 DOI: 10.2217/fon-2020-1290] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a common cancer globally and a leading cause of cancer-related deaths. Although early-stage disease may be curable by resection, liver transplantation or ablation, many patients present with unresectable disease and have a poor prognosis. Combination treatment with atezolizumab (targeting PD-L1) and bevacizumab (targeting VEGF) in the recent IMbrave150 study was shown to be effective with an acceptable safety profile in patients with unresectable HCC. Herein, we discuss this novel combination in the context of the liver immune environment, summarize the mechanism and pharmacokinetics of atezolizumab and bevacizumab, and examine recent data on other immune checkpoint inhibitor combination strategies as well as future directions in the treatment of patients with advanced HCC.
Collapse
Affiliation(s)
- Xiufeng Liu
- Department of Medical Oncology, Bayi Hospital Affiliated to Nanjing Chinese Medical University, Nanjing, 210002, China
| | - Yi Lu
- Shanghai Roche Pharmaceuticals Ltd, Shanghai, 201203, China
| | - Shukui Qin
- Department of Medical Oncology, Bayi Hospital Affiliated to Nanjing Chinese Medical University, Nanjing, 210002, China
| |
Collapse
|
13
|
Balza E, Carnemolla B, Orecchia P, Rubartelli A, Poggi A, Mortara L. Tumor Vasculature Targeted TNFα Therapy: Reversion of Microenvironment Anergy and Enhancement of the Anti-tumor Efficiency. Curr Med Chem 2020; 27:4233-4248. [PMID: 30182839 DOI: 10.2174/0929867325666180904121118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 05/16/2018] [Accepted: 07/18/2018] [Indexed: 02/07/2023]
Abstract
Tumor cells and tumor-associated stromal cells such as immune, endothelial and mesenchimal cells create a Tumor Microenvironment (TME) which allows tumor cell promotion, growth and dissemination while dampening the anti-tumor immune response. Efficient anti-tumor interventions have to keep into consideration the complexity of the TME and take advantage of immunotherapy and chemotherapy combined approaches. Thus, the aim of tumor therapy is to directly hit tumor cells and reverse endothelial and immune cell anergy. Selective targeting of tumor vasculature using TNFα-associated peptides or antibody fragments in association with chemotherapeutic agents, has been shown to exert a potent stimulatory effect on endothelial cells as well as on innate and adaptive immune responses. These drug combinations reducing the dose of single agents employed have led to minimize the associated side effects. In this review, we will analyze different TNFα-mediated tumor vesseltargeted therapies in both humans and tumor mouse models, with emphasis on the role played by the cross-talk between natural killer and dendritic cells and on the ability of TNFα to trigger tumor vessel activation and normalization. The improvement of the TNFα-based therapy with anti-angiogenic immunomodulatory drugs that may convert the TME from immunosuppressive to immunostimulant, will be discussed as well.
Collapse
Affiliation(s)
- Enrica Balza
- Cell Biology Unit, Ospedale Policlinico San Martino, Genoa, Italy
| | | | - Paola Orecchia
- Immunology Unit, Ospedale Policlinico San Martino, Genoa, Italy
| | - Anna Rubartelli
- Cell Biology Unit, Ospedale Policlinico San Martino, Genoa, Italy
| | - Alessandro Poggi
- Molecular Oncology and Angiogenesis Unit, IRCCS Policlinico San Martino, Genoa, Italy
| | - Lorenzo Mortara
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences (DBSV), University of Insubria, Via Monte Generoso, n. 71, 21100 Varese, Italy
| |
Collapse
|
14
|
Yang Y, Li L, Jiang Z, Wang B, Pan Z. Anlotinib optimizes anti-tumor innate immunity to potentiate the therapeutic effect of PD-1 blockade in lung cancer. Cancer Immunol Immunother 2020; 69:2523-2532. [PMID: 32577817 DOI: 10.1007/s00262-020-02641-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 06/16/2020] [Indexed: 12/30/2022]
Abstract
BACKGROUND Many anti-angiogenic agents have the potential to modulate the tumor microenvironment and improve immunotherapy. Anlotinib has demonstrated anti-tumor efficacy in non-small cell lung cancer (NSCLC) in third-line clinical trials. However, its roles in immune regulation and potentially synergistic anti-tumor effect in combination with immune checkpoint inhibition remain unclear. METHODS Here, based on a syngeneic lung cancer mouse model, the intratumoral immunological changes post-anlotinib treatment in the model were assessed. Furthermore, it was tested whether anlotinib could enhance the anti-tumor effect of αPD-1 in vivo. RESULTS This study shows that anlotinib increased infiltration of the innate immune cells, including natural killer (NK) cells, and antigen-presenting cells (APC), which include M1-like tumor-associated macrophages (TAM) and dendritic cells (DC), whereas the percentage of M2-like TAM was dramatically reduced. Subsequently, when combined with PD-1/PD-L1 (programmed cell death 1/PD-1 ligand 1) blockade, anlotinib conferred significantly synergistic therapeutic benefits. CONCLUSIONS Overall, these findings describe a role for anlotinib in the innate immune cells in the tumor microenvironment and a potentially synergistic anti-tumor combination with immune checkpoint inhibition.
Collapse
Affiliation(s)
- Yinli Yang
- Department of Integrative Oncology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.,Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Ling Li
- Department of Integrative Oncology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.,Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Zhansheng Jiang
- Department of Integrative Oncology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Bin Wang
- Department of Integrative Oncology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Zhanyu Pan
- Department of Integrative Oncology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China. .,Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China. .,Tianjin's Clinical Research Center for Cancer, Tianjin, China.
| |
Collapse
|
15
|
De Vito A, Orecchia P, Balza E, Reverberi D, Scaldaferri D, Taramelli R, Noonan DM, Acquati F, Mortara L. Overexpression of Murine Rnaset2 in a Colon Syngeneic Mouse Carcinoma Model Leads to Rebalance of Intra-Tumor M1/M2 Macrophage Ratio, Activation of T Cells, Delayed Tumor Growth, and Rejection. Cancers (Basel) 2020; 12:E717. [PMID: 32197460 PMCID: PMC7140044 DOI: 10.3390/cancers12030717] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/12/2020] [Accepted: 03/16/2020] [Indexed: 12/13/2022] Open
Abstract
Human RNASET2 acts as a powerful oncosuppressor protein in in vivo xenograft-based murine models of human cancer. Secretion of RNASET2 in the tumor microenvironment seems involved in tumor suppression, following recruitment of M1-polarized macrophages. Here, we report a murine Rnaset2-based syngeneic in vivo assay. BALB/c mice were injected with parental, empty vector-transfected or murine Rnaset2-overexpressing mouse C51 or TS/A syngeneic cells and tumor growth pattern and immune cells distribution in tumor mass were investigated. Compared to control cells, mouse Rnaset2-expressing C51 cells showed strong delayed tumor growth. CD86+ M1 macrophages were massively recruited in Rnaset2-expressing C51-derived tumors, with concomitant inhibition of MDSCs and CD206+ M2 macrophages recruitment. At later times, a relevant expansion of intra-tumor CD8+ T cells was also observed. After re-challenge with C51 parental cells, most mice previously injected with Rnaset2-expressing C51 cells still rejected C51 tumor cells, suggesting a Rnaset2-mediated T cell adaptive immune memory response. These results point at T2 RNases as evolutionary conserved oncosuppressors endowed with the ability to inhibit cancer growth in vivo through rebalance of intra-tumor M1/M2 macrophage ratio and concomitant recruitment of adaptive anti-tumor CD8+ T cells.
Collapse
Affiliation(s)
- Annarosaria De Vito
- Human Genetics Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (A.D.V.); (D.S.); (R.T.); (F.A.)
| | - Paola Orecchia
- Molecular Pathology Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy (D.R.)
| | - Enrica Balza
- Cell Biology Unit, IRCSS Ospedale Policlinico San Martino, 16132 Genova, Italy;
| | - Daniele Reverberi
- Molecular Pathology Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy (D.R.)
| | - Debora Scaldaferri
- Human Genetics Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (A.D.V.); (D.S.); (R.T.); (F.A.)
| | - Roberto Taramelli
- Human Genetics Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (A.D.V.); (D.S.); (R.T.); (F.A.)
| | - Douglas M. Noonan
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy;
- Scientific and Technology Pole, IRCCS MultiMedica, 20138 Milan, Italy
| | - Francesco Acquati
- Human Genetics Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (A.D.V.); (D.S.); (R.T.); (F.A.)
| | - Lorenzo Mortara
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy;
| |
Collapse
|
16
|
Bruno V, Corrado G, Baci D, Chiofalo B, Carosi MA, Ronchetti L, Piccione E, Albini A, Noonan DM, Piaggio G, Vizza E. Endometrial Cancer Immune Escape Mechanisms: Let Us Learn From the Fetal-Maternal Interface. Front Oncol 2020; 10:156. [PMID: 32226771 PMCID: PMC7080858 DOI: 10.3389/fonc.2020.00156] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 01/29/2020] [Indexed: 12/14/2022] Open
Abstract
The immune escape mechanisms at the base of tumor progression in endometrial cancer mimic immune tolerance mechanisms occurring at the maternal-fetal interface. The biological and immunological processes behind the maternal-fetal interface are finely tuned in time and space during embryo implantation and subsequent pregnancy stages; conversely, those behind cancer progression are often aberrant. The environment composition at the maternal-fetal interface parallels the pro-tumor microenvironment identified in many cancers, pointing to the possibility for the use of the maternal-fetal interface as a model to depict immune therapeutic targets in cancer. The framework of cancer environment signatures involved in immune adaptations, precisely timed in cancer progression, could reveal a specific "immune clock" in endometrial cancer, which might guide clinicians in patient risk class assessment, diagnostic workup, management, surgical and therapeutic approach, and surveillance strategies. Here, we review studies approaching this hypothesis, focusing on what is known so far about oncofetal similarities in immunity with the idea to individualize personalized immunotherapy targets, through the downregulation of the immune escape stage or the reactivation of the pro-inflammatory processes suppressed by the tumor.
Collapse
Affiliation(s)
- Valentina Bruno
- Gynecologic Oncology Unit, Department of Experimental Clinical Oncology, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Giacomo Corrado
- Gynecologic Oncology Unit, Department of Women and Children Health, Fondazione Policlinico Universitario A. Gemelli, IRCCS-Università Cattolica del Sacro Cuore, Rome, Italy
| | - Denisa Baci
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Benito Chiofalo
- Gynecologic Oncology Unit, Department of Experimental Clinical Oncology, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Maria Antonia Carosi
- Anatomy Pathology Unit, Department of Research, Diagnosis and Innovative Technologies, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Livia Ronchetti
- Anatomy Pathology Unit, Department of Research, Diagnosis and Innovative Technologies, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Emilio Piccione
- Section of Gynecology, Academic Department of Surgical Sciences, Tor Vergata University Hospital, University of Rome "Tor Vergata", Rome, Italy
| | - Adriana Albini
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.,Vascular Biology and Angiogenesis Laboratory, Science and Technology Pole (PST), IRCCS MultiMedica, Milan, Italy
| | - Douglas M Noonan
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy.,Vascular Biology and Angiogenesis Laboratory, Science and Technology Pole (PST), IRCCS MultiMedica, Milan, Italy
| | - Giulia Piaggio
- Department of Research, Diagnosis and Innovative Technologies, UOSD SAFU, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Enrico Vizza
- Gynecologic Oncology Unit, Department of Experimental Clinical Oncology, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
17
|
Huang X, Chen M, Wu H, Jiao Y, Zhou C. Macrophage Polarization Mediated by Chitooligosaccharide (COS) and Associated Osteogenic and Angiogenic Activities. ACS Biomater Sci Eng 2020; 6:1614-1629. [DOI: 10.1021/acsbiomaterials.9b01550] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Xiuhong Huang
- Department of Materials Science and Engineering, Jinan University, Guangzhou 510632, China
| | - Meng Chen
- Department of Materials Science and Engineering, Jinan University, Guangzhou 510632, China
| | - Haoming Wu
- Department of Materials Science and Engineering, Jinan University, Guangzhou 510632, China
| | - Yanpeng Jiao
- Department of Materials Science and Engineering, Jinan University, Guangzhou 510632, China
| | - Changren Zhou
- Department of Materials Science and Engineering, Jinan University, Guangzhou 510632, China
| |
Collapse
|
18
|
Grohé C, Gleiber W, Haas S, Losem C, Mueller-Huesmann H, Schulze M, Franke C, Basara N, Atz J, Kaiser R. Nintedanib plus docetaxel after progression on immune checkpoint inhibitor therapy: insights from VARGADO, a prospective study in patients with lung adenocarcinoma. Future Oncol 2019; 15:2699-2706. [DOI: 10.2217/fon-2019-0262] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Aim: To assess outcomes in patients with advanced adenocarcinoma non-small-cell lung cancer who received nintedanib plus docetaxel after progression on prior chemotherapy followed by immune checkpoint inhibitor (ICI) therapy. Patients & methods: VARGADO is a prospective, noninterventional study. We describe initial data from a cohort of 22 patients who received nintedanib plus docetaxel after chemotherapy and ICI therapy. Results: Median progression-free survival with nintedanib plus docetaxel was 5.5 months (95% CI: 1.9–8.7 months). The objective response rate was 7/12 (58%) and the disease control rate was 10/12 (83%). Data for overall survival rate 12 months after the start of treatment (primary end point) are not yet mature and are not reported. Of 22 patients, 73% experienced drug-related adverse events; adverse events led to treatment discontinuation in 32% of patients. Conclusion: These data highlight the potential clinical benefit of nintedanib plus docetaxel in patients who failed prior ICI therapy. Trial registration number: NCT02392455
Collapse
Affiliation(s)
- Christian Grohé
- Department of Respiratory Diseases, ELK, 13125, Berlin, Germany
| | - Wolfgang Gleiber
- University Hospital Frankfurt, Pneumology/Allergology, 60590 Frankfurt, Germany
| | - Siegfried Haas
- Friedrich-Ebert Hospital Neumuenster, Clinics for Hematology, Oncology & Nephrology, 24534 Neumuenster, Germany
| | - Christoph Losem
- MVZ für Onkologie und Hämatologie im Rhein-Kreis Neuss, 41462 Neuss, Germany
| | | | | | | | - Nadezda Basara
- Department of Internal Medicine, Malteser Hospital, St Franziskus, 24939 Flensburg, Germany
| | - Judith Atz
- Medical Affairs Oncology, Boehringer Ingelheim Pharma GmbH & Co KG, 55216 Ingelheim Am Rhein, Germany
| | - Rolf Kaiser
- Boehringer Ingelheim Pharma GmbH & Co, KG, 55216 Ingelheim Am Rhein, Germany
- Institute of Pharmacology, Johannes Gutenberg-University Mainz, 55122 Mainz, Germany
| |
Collapse
|
19
|
Qin H, Yu H, Sheng J, Zhang D, Shen N, Liu L, Tang Z, Chen X. PI3Kgamma Inhibitor Attenuates Immunosuppressive Effect of Poly(l-Glutamic Acid)-Combretastatin A4 Conjugate in Metastatic Breast Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1900327. [PMID: 31380170 PMCID: PMC6662090 DOI: 10.1002/advs.201900327] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 03/26/2019] [Indexed: 05/16/2023]
Abstract
Vascular disrupting agents (VDAs) have great potential for cancer treatment. Poly(l-glutamic acid)-combretastatin A4 conjugate (PLG-CA4) is a novel class of VDAs. Though it has notable antitumor activity, it can induce host immune responses that promote tumor growth. Here, PLG-CA4 induces the polarization of tumor-associated macrophages (TAMs) toward the M2-like phenotype in 4T1 metastatic breast cancer (Control 30% vs PLG-CA4 53%; p < 0.05). Compared to the monotherapy of PLG-CA4, inhibition of phosphoinositide 3-kinase gamma (PI3Kγ) attenuates the immunosuppressive effect of PLG-CA4 treatment by decreasing the number of M2-like TAMs (2.0 × 104 to 1.5 × 104 per tumor) and potential enhancement of cytotoxic T lymphocyte (3.0 × 104 to 5.7 × 104 per tumor). Importantly, PI3Kγ inhibitor synergizing with PLG-CA4 significantly extends the mean survival time from 52 days in monotherapy-treated mice to 61.8 days. Additionally, the combination of PLG-CA4 and PI3Kγ inhibitor improves the tumor therapeutic effect of NLG919, an inhibitor of immune checkpoint indoleamine 2,3-dioxygenase (IDO). As far as it is known, this is the first demonstrated study that VDAs induce the reshaping of macrophages to the M2-like phenotype. The findings also indicate a potential therapeutic strategy of the combination VDAs with an accurate immune modifier in the tumor to reverse the immune resistance.
Collapse
Affiliation(s)
- Hanjiao Qin
- Department of Radiotherapythe Second Hospital of Jilin UniversityChangchun130041P. R. China
| | - Haiyang Yu
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of SciencesChangchun130022P. R. China
- Jilin Biomedical Polymers Engineering LaboratoryChangchun130022P. R. China
| | - Jiyao Sheng
- Department of Hepatobiliary and Pancreatic Surgerythe Second Hospital of Jilin UniversityChangchun130041P. R. China
| | - Dawei Zhang
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of SciencesChangchun130022P. R. China
| | - Na Shen
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of SciencesChangchun130022P. R. China
- Jilin Biomedical Polymers Engineering LaboratoryChangchun130022P. R. China
| | - Linlin Liu
- Department of Radiotherapythe Second Hospital of Jilin UniversityChangchun130041P. R. China
| | - Zhaohui Tang
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of SciencesChangchun130022P. R. China
- Jilin Biomedical Polymers Engineering LaboratoryChangchun130022P. R. China
| | - Xuesi Chen
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of SciencesChangchun130022P. R. China
- Jilin Biomedical Polymers Engineering LaboratoryChangchun130022P. R. China
| |
Collapse
|
20
|
Gravis G, Billon E, Baldini C, Massard C, Hilgers W, Delva R, Walz J, Pignot G, Rybikowski S, Dermeche S, Thomassin J, Brunelle S, Lavaud P, Loriot Y. Unexpected response to cisplatin rechallenge after immune checkpoint inhibitors in patients with metastatic urothelial carcinoma refractory to platinum regimen. Eur J Cancer 2018; 104:236-238. [PMID: 30316610 DOI: 10.1016/j.ejca.2018.09.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 09/05/2018] [Accepted: 09/06/2018] [Indexed: 11/28/2022]
Affiliation(s)
- Gwenaëlle Gravis
- From the Centre de Recherche en Cancérologie de Marseille, INSERM UMR1068, CNRS UMR7258, Institut Paoli-Calmettes, Aix Marseille Université, Marseille, France; Medical Oncology, Institut Paoli-Calmettes, Marseille, France.
| | - Emilien Billon
- Medical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Capucine Baldini
- Drug Development Department (DITEP), Gustave Roussy, University of Paris Sud, Villejuif, France
| | - Christophe Massard
- Drug Development Department (DITEP), Gustave Roussy, University of Paris Sud, Villejuif, France
| | - Werner Hilgers
- Medical Oncology, Institut Sainte Catherine, Avignon, France
| | - Remy Delva
- Medical Oncology, Centre Paul Papin, Angers, France
| | - Jochen Walz
- Surgical Urology, Institut Paoli-Calmettes, Marseille, France
| | | | | | | | - Jeanne Thomassin
- Biopathology Department Institut Paoli-Calmettes, Marseille, France
| | - Serge Brunelle
- Radiology Department Institut Paoli-Calmettes, Marseille, France
| | - Pernelle Lavaud
- Département de Médecine Oncologique, Gustave Roussy, University of Paris Sud, Villejuif, France
| | - Yohann Loriot
- Département de Médecine Oncologique, Gustave Roussy, University of Paris Sud, Villejuif, France
| | | |
Collapse
|
21
|
Bates DO, Beazley-Long N, Benest AV, Ye X, Ved N, Hulse RP, Barratt S, Machado MJ, Donaldson LF, Harper SJ, Peiris-Pages M, Tortonese DJ, Oltean S, Foster RR. Physiological Role of Vascular Endothelial Growth Factors as Homeostatic Regulators. Compr Physiol 2018; 8:955-979. [PMID: 29978898 DOI: 10.1002/cphy.c170015] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The vascular endothelial growth factor (VEGF) family of proteins are key regulators of physiological systems. Originally linked with endothelial function, they have since become understood to be principal regulators of multiple tissues, both through their actions on vascular cells, but also through direct actions on other tissue types, including epithelial cells, neurons, and the immune system. The complexity of the five members of the gene family in terms of their different splice isoforms, differential translation, and specific localizations have enabled tissues to use these potent signaling molecules to control how they function to maintain their environment. This homeostatic function of VEGFs has been less intensely studied than their involvement in disease processes, development, and reproduction, but they still play a substantial and significant role in healthy control of blood volume and pressure, interstitial volume and drainage, renal and lung function, immunity, and signal processing in the peripheral and central nervous system. The widespread expression of VEGFs in healthy adult tissues, and the disturbances seen when VEGF signaling is inhibited support this view of the proteins as endogenous regulators of normal physiological function. This review summarizes the evidence and recent breakthroughs in understanding of the physiology that is regulated by VEGF, with emphasis on the role they play in maintaining homeostasis. © 2017 American Physiological Society. Compr Physiol 8:955-979, 2018.
Collapse
Affiliation(s)
- David O Bates
- Cancer Biology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom
| | | | - Andrew V Benest
- Cancer Biology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom
| | - Xi Ye
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Nikita Ved
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Richard P Hulse
- Cancer Biology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom
| | - Shaney Barratt
- Academic Respiratory Unit, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| | - Maria J Machado
- Cancer Biology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom
| | - Lucy F Donaldson
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Steven J Harper
- School of Physiology, Pharmacology & Neuroscience, Medical School, University of Bristol, Bristol, United Kingdom
| | - Maria Peiris-Pages
- Cancer Research UK Manchester Institute, The University of Manchester, Manchester, United Kingdom
| | - Domingo J Tortonese
- Centre for Comparative and Clinical Anatomy, University of Bristol, Bristol, United Kingdom
| | - Sebastian Oltean
- Institute of Biomedical & Clinical Sciences, University of Exeter Medical School, Exeter, United Kingdom
| | - Rebecca R Foster
- Bristol Renal, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
22
|
Bruno A, Bassani B, D'Urso DG, Pitaku I, Cassinotti E, Pelosi G, Boni L, Dominioni L, Noonan DM, Mortara L, Albini A. Angiogenin and the MMP9-TIMP2 axis are up-regulated in proangiogenic, decidual NK-like cells from patients with colorectal cancer. FASEB J 2018; 32:5365-5377. [PMID: 29763380 DOI: 10.1096/fj.201701103r] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
NK cells are effector lymphocytes involved in tumor immunosurveillance; however, in patients with solid malignancies, NK cells have compromised functions. We have previously reported that lung tumor-associated NK cells (TANKs; peripheral blood) and tumor-infiltrating NK cells (TINKs) show proangiogenic, decidual NK-like (dNK) phenotype. In this study, we functionally and molecularly investigated TINKs and TANKs from blood and tissue samples of patients with colorectal cancer (CRC), a neoplasm in which inflammation and angiogenesis have clinical relevance, and compared them to NK cells from controls and patients with nononcologic inflammatory bowel disease. CRC TINKs/TANKs showed decreased expression for the activatory marker NKG2D, impaired degranulation activity, a decidual-like NK polarization toward the CD56brightCD16dim/-CD9+CD49+ subset. TINKs and TANKs secreted cytokines with proangiogenic activities, and induce endothelial cell proliferation, migration, adhesion, and the formation of capillary-like structures in vitro. dNK cells release specific proangiogenic factors; among which, angiogenin and invasion-associated enzymes related to the MMP9-TIMP1/2 axis. Here, we describe, for the first time, to our knowledge, the expression of angiogenin, MMP2/9, and TIMP by TANKs in patients with CRC. This phenotype could be relevant to the invasive capabilities and proangiogenic functions of CRC-NK cells and become a novel biomarker. STAT3/STAT5 activation was observed in CRC-TANKs, and treatment with pimozide, a STAT5 inhibitor, reduced endothelial cell capability to form capillary-like networks, inhibiting VEGF and angiogenin production without affecting the levels of TIMP1, TIMP2, and MMP9, indicating that STAT5 is involved in cytokine modulation but not invasion-associated molecules. Combination of Stat5 or MMP inhibitors with immunotherapy could help repolarize CRC TINKs and TANKs to anti-tumor antimetastatic ones.-Bruno, A., Bassani, B., D'Urso, D. G., Pitaku, I., Cassinotti, E., Pelosi, G., Boni, L., Dominioni, L., Noonan, D. M., Mortara, L., Albini, A. Angiogenin and the MMP9-TIMP2 axis are up-regulated in proangiogenic, decidual NK-like cells from patients with colorectal cancer.
Collapse
Affiliation(s)
- Antonino Bruno
- Vascular Biology and Angiogenesis Laboratory, Science and Technology Pole (PST), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Milan, Italy
| | - Barbara Bassani
- Vascular Biology and Angiogenesis Laboratory, Science and Technology Pole (PST), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Milan, Italy
| | - Davide Giuseppe D'Urso
- Vascular Biology and Angiogenesis Laboratory, Science and Technology Pole (PST), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Milan, Italy
| | - Ilvana Pitaku
- Department of Surgical and Morphological Sciences, University of Insubria, Varese, Italy
| | - Elisa Cassinotti
- Department of Surgical and Morphological Sciences, University of Insubria, Varese, Italy
| | - Giuseppe Pelosi
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Luigi Boni
- Department of Medical-Surgical Physiopathology and Transplantation, University of Milan-Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Ca' Granda, Polyclinic Hospital, Milan, Italy
| | - Lorenzo Dominioni
- Department of Surgical and Morphological Sciences, University of Insubria, Varese, Italy
| | - Douglas M Noonan
- Vascular Biology and Angiogenesis Laboratory, Science and Technology Pole (PST), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Milan, Italy.,Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy; and
| | - Lorenzo Mortara
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy; and
| | - Adriana Albini
- Vascular Biology and Angiogenesis Laboratory, Science and Technology Pole (PST), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Milan, Italy.,Department of Medicine and Surgery, University Milano-Bicocca, Monza, Italy
| |
Collapse
|
23
|
Albini A, Bruno A, Noonan DM, Mortara L. Contribution to Tumor Angiogenesis From Innate Immune Cells Within the Tumor Microenvironment: Implications for Immunotherapy. Front Immunol 2018; 9:527. [PMID: 29675018 PMCID: PMC5895776 DOI: 10.3389/fimmu.2018.00527] [Citation(s) in RCA: 289] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 02/28/2018] [Indexed: 12/14/2022] Open
Abstract
The critical role of angiogenesis in promoting tumor growth and metastasis is strongly established. However, tumors show considerable variation in angiogenic characteristics and in their sensitivity to antiangiogenic therapy. Tumor angiogenesis involves not only cancer cells but also various tumor-associated leukocytes (TALs) and stromal cells. TALs produce chemokines, cytokines, proteases, structural proteins, and microvescicles. Vascular endothelial growth factor (VEGF) and inflammatory chemokines are not only major proangiogenic factors but are also immune modulators, which increase angiogenesis and lead to immune suppression. In our review, we discuss the regulation of angiogenesis by innate immune cells in the tumor microenvironment, specific features, and roles of major players: macrophages, neutrophils, myeloid-derived suppressor and dendritic cells, mast cells, γδT cells, innate lymphoid cells, and natural killer cells. Anti-VEGF or anti-inflammatory drugs could balance an immunosuppressive microenvironment to an immune permissive one. Anti-VEGF as well as anti-inflammatory drugs could therefore represent partners for combinations with immune checkpoint inhibitors, enhancing the effects of immune therapy.
Collapse
Affiliation(s)
- Adriana Albini
- Scientific and Technology Pole, IRCCS MultiMedica, Milano, Italy.,Department of Medicine and Surgery, University Milano-Bicocca, Monza, Italy
| | - Antonino Bruno
- Scientific and Technology Pole, IRCCS MultiMedica, Milano, Italy
| | - Douglas M Noonan
- Scientific and Technology Pole, IRCCS MultiMedica, Milano, Italy.,Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Lorenzo Mortara
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| |
Collapse
|
24
|
Macrophage Polarization in Chronic Inflammatory Diseases: Killers or Builders? J Immunol Res 2018. [PMID: 29507865 DOI: 10.1155/2018/8917804]] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Macrophages are key cellular components of the innate immunity, acting as the main player in the first-line defence against the pathogens and modulating homeostatic and inflammatory responses. Plasticity is a major feature of macrophages resulting in extreme heterogeneity both in normal and in pathological conditions. Macrophages are not homogenous, and they are generally categorized into two broad but distinct subsets as either classically activated (M1) or alternatively activated (M2). However, macrophages represent a continuum of highly plastic effector cells, resembling a spectrum of diverse phenotype states. Induction of specific macrophage functions is closely related to the surrounding environment that acts as a relevant orchestrator of macrophage functions. This phenomenon, termed polarization, results from cell/cell, cell/molecule interaction, governing macrophage functionality within the hosting tissues. Here, we summarized relevant cellular and molecular mechanisms driving macrophage polarization in "distant" pathological conditions, such as cancer, type 2 diabetes, atherosclerosis, and periodontitis that share macrophage-driven inflammation as a key feature, playing their dual role as killers (M1-like) and/or builders (M2-like). We also dissect the physio/pathological consequences related to macrophage polarization within selected chronic inflammatory diseases, placing polarized macrophages as a relevant hallmark, putative biomarkers, and possible target for prevention/therapy.
Collapse
|
25
|
Macrophage Polarization in Chronic Inflammatory Diseases: Killers or Builders? J Immunol Res 2018; 2018:8917804. [PMID: 29507865 PMCID: PMC5821995 DOI: 10.1155/2018/8917804] [Citation(s) in RCA: 312] [Impact Index Per Article: 44.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Revised: 11/01/2017] [Accepted: 11/15/2017] [Indexed: 12/13/2022] Open
Abstract
Macrophages are key cellular components of the innate immunity, acting as the main player in the first-line defence against the pathogens and modulating homeostatic and inflammatory responses. Plasticity is a major feature of macrophages resulting in extreme heterogeneity both in normal and in pathological conditions. Macrophages are not homogenous, and they are generally categorized into two broad but distinct subsets as either classically activated (M1) or alternatively activated (M2). However, macrophages represent a continuum of highly plastic effector cells, resembling a spectrum of diverse phenotype states. Induction of specific macrophage functions is closely related to the surrounding environment that acts as a relevant orchestrator of macrophage functions. This phenomenon, termed polarization, results from cell/cell, cell/molecule interaction, governing macrophage functionality within the hosting tissues. Here, we summarized relevant cellular and molecular mechanisms driving macrophage polarization in “distant” pathological conditions, such as cancer, type 2 diabetes, atherosclerosis, and periodontitis that share macrophage-driven inflammation as a key feature, playing their dual role as killers (M1-like) and/or builders (M2-like). We also dissect the physio/pathological consequences related to macrophage polarization within selected chronic inflammatory diseases, placing polarized macrophages as a relevant hallmark, putative biomarkers, and possible target for prevention/therapy.
Collapse
|