1
|
Sunitinib Combined with Th1 Cytokines Potentiates Apoptosis in Human Breast Cancer Cells and Suppresses Tumor Growth in a Murine Model of HER-2 pos Breast Cancer. Int J Breast Cancer 2021; 2021:8818393. [PMID: 33936816 PMCID: PMC8062178 DOI: 10.1155/2021/8818393] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 04/03/2021] [Indexed: 11/18/2022] Open
Abstract
Although immune-based therapies have made remarkable inroads in cancer treatment, they usually must be combined with standard treatment modalities, including cytotoxic drugs, to achieve maximal clinical benefits. As immunotherapies are further advanced and refined, considerable efforts will be required to identify combination therapies that will maximize clinical responses while simultaneously decreasing the unpleasant and sometimes life-threatening side effects of standard therapy. Over the last two decades, evidence has emerged that Th1 cytokines can play a central role in protective antitumor immunity and that combinations of Th1 cytokines can induce senescence and apoptosis in cancer cells. To explore the possibility of combining targeted drugs with Th1-polarizing vaccines, we undertook a study to examine the impact of combining Th1 cytokines with the relatively broad-spectrum receptor tyrosine kinase antagonist, sunitinib. We found that when a panel of five phenotypically diverse human breast cancer cell lines was subjected to treatment with sunitinib plus recombinant Th1 cytokines IFN-γ and TNF-α, synergistic effects were observed across a number of parameters including different aspects of apoptotic cell death. Interestingly, sunitinib was found to have a profoundly suppressive effect of T cell's capacity to secrete IFN-γ, indicating that in vivo use of this drug may hinder robust Th1 responses. Nonetheless, this suppression was circumvented in a mouse model of HER-2pos breast disease by supplying recombinant interferon-gamma to achieve a combination therapy significantly more potent than either agent.
Collapse
|
2
|
Cimadamore A, Massari F, Santoni M, Lopez-Beltran A, Cheng L, Scarpelli M, Montironi R, Moch H. PD1 and PD-L1 Inhibitors for the Treatment of Kidney Cancer: The Role of PD-L1 Assay. Curr Drug Targets 2020; 21:1664-1671. [PMID: 32208115 DOI: 10.2174/1389450121666200324151056] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 02/28/2020] [Accepted: 03/02/2020] [Indexed: 01/11/2023]
Abstract
BACKGROUND Immune checkpoint inhibitors targeting the programmed death receptor ligand 1 (PD-L1)/programmed death receptor 1 (PD-1) pathway represent a drastic change in the treatment landscape of RCC resulting in a dynamic and evolving scenario. There is an urgent need for predictive biomarkers of response to provide a personalized therapeutic strategy for individual patients. OBJECTIVE In this review, we focused on trials that investigated the administration of a PD-1 and PDL1 inhibitor alone or in combination with another agent and compared the different assays applied in each trial to evaluate the role of PD-L1 as a prognostic and predictive biomarker. CONCLUSION So far, the use of PD-L1 expression alone is not sufficient to predict treatment response and present many limitations: the lack of consensus between different methodologies on biomarker assessment, the heterogeneity of PD-L1 between primary tumors and metastatic sites, different criteria of response to therapy (RECIST vs. irRECIST), the complex interplay with inflammatory components, previous treatments, administration of antibiotic therapy. Combinations of different biomarkers and biological features, such as gene expression associated with angiogenesis, immune response and myeloid inflammation are promising biological variables that need to be validated in the context of prospective clinical trials.
Collapse
Affiliation(s)
- Alessia Cimadamore
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| | | | | | - Antonio Lopez-Beltran
- Department of Pathology and Surgery, University of Cordoba, Faculty of Medicine, Cordoba, Spain
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Marina Scarpelli
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| | - Rodolfo Montironi
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| | - Holger Moch
- Department of Pathology and Molecular Pathology, University and University Hospital Zurich, CH-8091 Zurich, Switzerland
| |
Collapse
|
3
|
Ruiz-Camps I, Aguilar-Company J. Epidermal Growth Factor Receptor Inhibitors and Other Tyrosine Kinase Inhibitors for Solid Tumors. Infect Dis Clin North Am 2020; 34:257-270. [PMID: 32334988 DOI: 10.1016/j.idc.2020.02.005] [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: 10/24/2022]
Abstract
This article analyzes the risk of infection associated with small molecule kinase inhibitors used to treat solid organ malignancies and establishes specific recommendations. Most of these drugs are orally administered and have the ability to inhibit distinct kinases, which play a major role in cancer initiation and progression. Although the true extent of adverse events is not yet known, risk of infection does not seem to be a major problem with these drugs. Because of the limited clinical experience and the constant evolution of targeted therapies, recommendations may evolve in the near future.
Collapse
Affiliation(s)
- Isabel Ruiz-Camps
- Infectious Diseases Department, Vall d'Hebron University Hospital, Passeig de la Vall d'Hebron, 119-129, Barcelona 08035, Spain
| | - Juan Aguilar-Company
- Infectious Diseases Department, Vall d'Hebron University Hospital, Passeig de la Vall d'Hebron, 119-129, Barcelona 08035, Spain; Oncology Department, Vall d'Hebron University Hospital, Passeig de la Vall d'Hebron, 119-129, Barcelona 08035, Spain.
| |
Collapse
|
4
|
Lopez-Beltran A, Henriques V, Cimadamore A, Santoni M, Cheng L, Gevaert T, Blanca A, Massari F, Scarpelli M, Montironi R. The Identification of Immunological Biomarkers in Kidney Cancers. Front Oncol 2018; 8:456. [PMID: 30450335 PMCID: PMC6225533 DOI: 10.3389/fonc.2018.00456] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 10/01/2018] [Indexed: 12/18/2022] Open
Abstract
The recent approval of several agents have revolutionized the scenario of therapeutic management of metastatic renal cell carcinoma (RCC) allowing us to reach important clinical end points with extended patients' survival. Actually, every new drug approved has represented an important step forward to the improvement of patient's survival. On the other hand, we now understand that RCC includes a large group of tumor entities, each of them with different genetic and mutational alterations, but also showing different clinical behavior; a reason behind the needs of subtype specific personalized approach to therapy of RCC. Immunotherapy is gradually becoming a key factor in the therapeutic algorithm for patients with locally advanced or metastatic RCC. Due to the combination of potent treatment success and potentially deadly adverse effects from immune checkpoint inhibitors (ICI), gathering prognostic and predictive information about FDA-indicated tumors seems to be prudent. Robust and reliable biomarkers are crucial for patient's selection of treatments with immunomodulatory drugs. PD-L1 expression is a poor prognostic factor and predictive of better responses from both PD-1 and PD-L1 inhibitors in a variety of tumor types including RCC. Each FDA approved PD-1/PD-L1 drug is paired with a PD-L1 Immunohistochemistry (IHC) assay. Thus, there is need for improved knowledge and application of PD-1/PD-L1 IHC biomarkers in daily practice. IHC staining appears in membranous fashion. The atezolizumab approved IHC assay is unique in that only immune cell staining is quantified for the use of this assay in RCC. A single biomarker for patient selection may not be feasible, given that immune responses are dynamic and evolve over time. Biomarker development for ICI drugs will likely require integration of multiple biologic components like PD-L1 expression, TILs and mutational load. New methodological approaches based on digital pathology may be relevant since they will allow recognition of the biomarker and to objectively quantitate its expression, and therefore might produce objective and reproducible cut-off assessment. Multidisciplinary approach is very much needed to fully develop the current and future value of ICI in clinical practice.
Collapse
Affiliation(s)
- Antonio Lopez-Beltran
- Department of Pathology and Surgery, Faculty of Medicine, Cordoba University, Cordoba, Spain
| | | | - Alessia Cimadamore
- Section of Pathological Anatomy, United Hospital, School of Medicine, Polytechnic University of the Marche Region, Ancona, Italy
| | | | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Thomas Gevaert
- Laboratory of Experimental Urology, Organ Systems, KU Leuven, Leuven, Belgium.,Department of Pathology, AZ Klina, Brasschaat, Belgium
| | - Ana Blanca
- Instituto Maimonides de Investigación Biomédica de Córdoba, Córdoba, Spain
| | | | - Marina Scarpelli
- Section of Pathological Anatomy, United Hospital, School of Medicine, Polytechnic University of the Marche Region, Ancona, Italy
| | - Rodolfo Montironi
- Section of Pathological Anatomy, United Hospital, School of Medicine, Polytechnic University of the Marche Region, Ancona, Italy
| |
Collapse
|
5
|
Santoni M, Massari F, Di Nunno V, Conti A, Cimadamore A, Scarpelli M, Montironi R, Cheng L, Battelli N, Lopez-Beltran A. Immunotherapy in renal cell carcinoma: latest evidence and clinical implications. Drugs Context 2018; 7:212528. [PMID: 29899754 PMCID: PMC5992965 DOI: 10.7573/dic.212528] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 04/11/2018] [Accepted: 04/16/2018] [Indexed: 12/12/2022] Open
Abstract
Advances in understanding the mechanisms of tumour-induced immunosuppression have led to the development of immune-checkpoint inhibitors in cancer patients, including those with renal cell carcinoma (RCC). The optimal combination between immunotherapy and targeted agents (as well as the possible favourable sequential therapy of these two classes of drugs) remains an open question at this moment. Several trials are currently underway to assess the combination of anti-programmed-death 1 (PD-1) or anti-PD-ligand(L)1 agents with other immunotherapies or with anti-vascular endothelial growth factor receptor (VEGFR) tyrosine kinase inhibitors (TKIs). In this editorial, we described the results of the most recent clinical trials on the use of immunotherapies in RCC and the emerging data on the research for reliable biomarkers of tumour response in this setting. In addition, we have focused on the role of the gut microbiome and tumour microenvironment in the development of future therapeutic strategies for RCC patients.
Collapse
Affiliation(s)
| | | | | | - Alessandro Conti
- Department of Urology, Bressanone/Brixen Hospital, Bressanone, Italy
| | - Alessia Cimadamore
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| | - Marina Scarpelli
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| | - Rodolfo Montironi
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Antonio Lopez-Beltran
- Department of Pathology and Surgery, University of Cordoba, Faculty of Medicine, Cordoba, Spain
| |
Collapse
|
6
|
Noé G, Bellesoeur A, Thomas-Schoemann A, Rangarajan S, Naji F, Puszkiel A, Huillard O, Saidu N, Golmard L, Alexandre J, Goldwasser F, Blanchet B, Vidal M. Clinical and kinomic analysis identifies peripheral blood mononuclear cells as a potential pharmacodynamic biomarker in metastatic renal cell carcinoma patients treated with sunitinib. Oncotarget 2018; 7:67507-67520. [PMID: 27589830 PMCID: PMC5341893 DOI: 10.18632/oncotarget.11686] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 07/10/2016] [Indexed: 12/31/2022] Open
Abstract
Background Sunitinib is a protein tyrosine kinase (PTK) inhibitor that has immune-modulating properties. In this context, peripheral blood mononuclear cells (PBMC), mainly constituted by lymphocytes, could be a perfect surrogate tissue for identifying and assaying pharmacodynamic biomarkers of sunitinib. In this study, we investigated the changes in lymphocytes count as pharmacodynamic biomarker in metastatic renal cell carcinoma (mRCC) patients under sunitinib therapy. Thereafter, we studied the ex vivo effect of sunitinib and SU12262 (active metabolite) on PBMC from naïve mRCC patients using a high throughput kinomic profiling method. Methods The prognostic value of total lymphocytes count between Day 0 and Day 21 (expressed as a ratio D21/D0) was retrospectively investigated in 88 mRCC patients under sunitinib therapy. PTK PamChip® microarrays were used to explore prospectively the ex vivo effect of sunitinib and SU12662 on PTK activity in PBMC from 21 naïve mRCC patients. Results In this retrospective study, D21/D0 lymphocytes ratio (Hazard Ratio, 1.83; CI95%, 1.24-2.71; p=0.0023) was independently associated with PFS. Interestingly, kinomic analysis showed that D21/D0 lymphocytes ratio and Heng prognostic model was statistically associated with the ex vivo sunitinib and SU12662 effect in PBMC. Conclusion The present study highlights that D21/D0 total lymphocytes ratio could be a promising pharmacodynamic biomarker in mRCC patients treated with sunitinib. Additionally, it paves the way to investigate the kinomic profile in PBMC as a prognostic factor in a larger cohort of mRCC patients under sunitinib therapy.
Collapse
Affiliation(s)
- Gaёlle Noé
- Assistance Publique Hôpitaux de Paris, Hôpital Cochin, UF Pharmacocinétique et Pharmacochimie, Paris, France.,UMR8638 CNRS, Faculté de Pharmacie, Université Paris Descartes, PRES Sorbonne Paris Cité, Paris, France
| | - Audrey Bellesoeur
- Assistance Publique Hôpitaux de Paris, Hôpital Cochin, UF Pharmacocinétique et Pharmacochimie, Paris, France.,UMR8638 CNRS, Faculté de Pharmacie, Université Paris Descartes, PRES Sorbonne Paris Cité, Paris, France.,Assistance Publique Hôpitaux de Paris, Hôpital Cochin, Service de Cancérologie Médicale, Paris, France
| | - Audrey Thomas-Schoemann
- Assistance Publique Hôpitaux de Paris, Hôpital Cochin, UF Pharmacocinétique et Pharmacochimie, Paris, France.,UMR8638 CNRS, Faculté de Pharmacie, Université Paris Descartes, PRES Sorbonne Paris Cité, Paris, France
| | | | - Faris Naji
- PamGene International BV, 's-Hertogenbosch, The Netherlands
| | - Alicja Puszkiel
- Assistance Publique Hôpitaux de Paris, Hôpital Cochin, UF Pharmacocinétique et Pharmacochimie, Paris, France
| | - Olivier Huillard
- Assistance Publique Hôpitaux de Paris, Hôpital Cochin, Service de Cancérologie Médicale, Paris, France
| | - Nathaniel Saidu
- U1016 INSERM, UMR 8104 CNRS, UMR-S1016, CARPEM, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Lisa Golmard
- Institut Curie, Département de Biopathologie, Paris, France
| | - Jerome Alexandre
- Assistance Publique Hôpitaux de Paris, Hôpital Cochin, Service de Cancérologie Médicale, Paris, France.,U1016 INSERM, UMR 8104 CNRS, UMR-S1016, CARPEM, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Francois Goldwasser
- Assistance Publique Hôpitaux de Paris, Hôpital Cochin, Service de Cancérologie Médicale, Paris, France.,U1016 INSERM, UMR 8104 CNRS, UMR-S1016, CARPEM, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Benoit Blanchet
- Assistance Publique Hôpitaux de Paris, Hôpital Cochin, UF Pharmacocinétique et Pharmacochimie, Paris, France
| | - Michel Vidal
- Assistance Publique Hôpitaux de Paris, Hôpital Cochin, UF Pharmacocinétique et Pharmacochimie, Paris, France.,UMR8638 CNRS, Faculté de Pharmacie, Université Paris Descartes, PRES Sorbonne Paris Cité, Paris, France
| |
Collapse
|
7
|
Aguilar-Company J, Fernández-Ruiz M, García-Campelo R, Garrido-Castro AC, Ruiz-Camps I. ESCMID Study Group for Infections in Compromised Hosts (ESGICH) Consensus Document on the safety of targeted and biological therapies: an infectious diseases perspective (Cell surface receptors and associated signaling pathways). Clin Microbiol Infect 2018; 24 Suppl 2:S41-S52. [PMID: 29426804 DOI: 10.1016/j.cmi.2017.12.027] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 12/18/2017] [Accepted: 12/30/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND The present review is part of the European Society of Clinical Microbiology and Infectious Diseases (ESCMID) Study Group for Infections in Compromised Hosts (ESGICH) consensus document on the safety of targeted and biologic therapies. AIMS To review, from an infectious diseases perspective, the safety profile of therapies targeting cell surface receptors and associated signaling pathways among cancer patients and to suggest preventive recommendations. SOURCES Computer-based Medline searches with MeSH terms pertaining to each agent or therapeutic family. CONTENT Vascular endothelial growth factor (VEGF)-targeted agents (bevacizumab and aflibercept) are associated with a meaningful increase in the risk of infection, likely due to drug-induced neutropaenia, although no clear benefit is expected from the universal use of anti-infective prophylaxis. VEGF tyrosine kinase inhibitors (i.e. sorafenib or sunitinib) do not seem to significantly affect host's susceptibility to infection, and universal anti-infective prophylaxis is not recommended either. Anti-epidermal growth factor receptor (EGFR) monoclonal antibodies (cetuximab or panitumumab) induce neutropaenia and secondary skin and soft tissue infection in cases of severe papulopustular rash. Systemic antibiotics (doxycycline or minocycline) should be administered to prevent the latter complication, whereas no recommendation can be established on the benefit from antiviral, antifungal or anti-Pneumocystis prophylaxis. A lower risk of infection is reported for anti-ErbB2/HER2 monoclonal antibodies (trastuzumab and pertuzumab) and ErbB receptor tyrosine kinase inhibitors (including dual-EGFR/ErbB2 inhibitors such as lapatinib or neratinib) compared to conventional chemotherapy, presumably as a result of the decreased occurrence of drug-induced neutropaenia. IMPLICATIONS With the exception of VEGF-targeted agents, the overall risk of infection associated with the reviewed therapies seems to be low.
Collapse
Affiliation(s)
- J Aguilar-Company
- Departments of Infectious Diseases and Oncology, University Hospital Vall d'Hebron, Autonomous University of Barcelona, Barcelona, Spain
| | - M Fernández-Ruiz
- Unit of Infectious Diseases, Hospital Universitario '12 de Octubre', Instituto de Investigación Hospital '12 de Octubre' (i + 12), School of Medicine, Universidad Complutense, Madrid, Spain; Spanish Network for Research in Infectious Diseases (REIPI RD16/0016), Instituto de Salud Carlos III, Madrid, Spain
| | - R García-Campelo
- Department of Medical Oncology, Complejo Hospitalario Universitario A Coruña, A Coruña, Spain
| | - A C Garrido-Castro
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - I Ruiz-Camps
- Department of Infectious Diseases, University Hospital Vall d'Hebron, Autonomous University of Barcelona, Barcelona, Spain; Spanish Network for Research in Infectious Diseases (REIPI RD16/0016), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
8
|
Moeckel J, Staiger N, Mackensen A, Meidenbauer N, Ullrich E. Sunitinib does not impair natural killer cell function in patients with renal cell carcinoma. Oncol Lett 2017; 14:1089-1096. [PMID: 28693278 PMCID: PMC5494766 DOI: 10.3892/ol.2017.6187] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 11/10/2016] [Indexed: 01/17/2023] Open
Abstract
Although the available treatment options have expanded, the survival of patients with metastatic renal cell carcinoma (RCC) remains poor. As patients with RCC lack responsiveness to chemotherapy or radiation, therapeutic options predominantly include surgical interventions and immunomodulatory approaches, including the administration of tyrosine kinase inhibitors (TKIs) such as sunitinib. Natural killer (NK) cells have been reported to be key players in TKI-mediated off-target effects on the immune system. However, only limited information is available regarding the possible impact of sunitinib on the function of NK cells of individual patients. The present study reports on the immunomonitoring results of three patients with metastatic RCC who underwent sunitinib treatment. These results were compared with age-matched, healthy controls in terms of the immune status of T, B and NK cells, focusing on functional in vitro analyses of NK cells. In all three patients, NK cell number, subset distribution and function, as measured by cluster of differentiation 107a degranulation, did not exhibit any significant alterations as a result of sunitinib treatment. These results indicate that sunitinib does not negatively affect NK cell function, which supports the pursuit of therapeutic modalities that combine immunomodulation and NK cell-stimulating approaches.
Collapse
Affiliation(s)
- Jennifer Moeckel
- Department of Internal Medicine 5, Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, D-91054 Erlangen, Germany
- LOEWE Center for Cell and Gene Therapy, Goethe University Frankfurt, D-60590 Frankfurt, Germany
| | - Nina Staiger
- LOEWE Center for Cell and Gene Therapy, Goethe University Frankfurt, D-60590 Frankfurt, Germany
- Laboratory of Cellular Immunology, Division for Stem Cell Transplantation and Immunology, Hospital for Children and Adolescents, Goethe University Frankfurt, D-60590 Frankfurt, Germany
| | - Andreas Mackensen
- Department of Internal Medicine 5, Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, D-91054 Erlangen, Germany
| | - Norbert Meidenbauer
- Department of Internal Medicine 5, Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, D-91054 Erlangen, Germany
| | - Evelyn Ullrich
- Department of Internal Medicine 5, Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, D-91054 Erlangen, Germany
- LOEWE Center for Cell and Gene Therapy, Goethe University Frankfurt, D-60590 Frankfurt, Germany
- Laboratory of Cellular Immunology, Division for Stem Cell Transplantation and Immunology, Hospital for Children and Adolescents, Goethe University Frankfurt, D-60590 Frankfurt, Germany
| |
Collapse
|
9
|
Reinwald M, Boch T, Hofmann WK, Buchheidt D. Risk of Infectious Complications in Hemato-Oncological Patients Treated with Kinase Inhibitors. Biomark Insights 2016; 10:55-68. [PMID: 27127405 PMCID: PMC4841329 DOI: 10.4137/bmi.s22430] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 09/13/2015] [Accepted: 09/15/2015] [Indexed: 12/30/2022] Open
Abstract
Infectious complications are a major cause of morbidity and mortality in patients with hemato-oncological diseases. Although disease-related immunosuppression represents one factor, aggressive treatment regimens, such as chemotherapy, stem cell transplantation, or antibody treatment, account for a large proportion of infectious side effects. With the advent of targeted therapies affecting specific kinases in malignant diseases, the outcome of patients has further improved. Nonetheless, dependent on the specific pathway targeted or off-target activity of the kinase inhibitor, therapy-associated infectious complications may occur. We review the most common and approved kinase inhibitors targeting a variety of hemato-oncological malignancies for their immunosuppressive potential and evaluate their risk of infectious side effects based on preclinical evidence and clinical data in order to raise awareness of the potential risks involved.
Collapse
Affiliation(s)
- Mark Reinwald
- Department of Hematology and Oncology, Mannheim University Hospital, University of Heidelberg, Mannheim, Germany
| | - Tobias Boch
- Department of Hematology and Oncology, Mannheim University Hospital, University of Heidelberg, Mannheim, Germany
| | - Wolf-Karsten Hofmann
- Department of Hematology and Oncology, Mannheim University Hospital, University of Heidelberg, Mannheim, Germany
| | - Dieter Buchheidt
- Department of Hematology and Oncology, Mannheim University Hospital, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
10
|
Santoni M, Berardi R, Amantini C, Burattini L, Santini D, Santoni G, Cascinu S. Role of natural and adaptive immunity in renal cell carcinoma response to VEGFR-TKIs and mTOR inhibitor. Int J Cancer 2013; 134:2772-7. [PMID: 24114790 DOI: 10.1002/ijc.28503] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Accepted: 09/12/2013] [Indexed: 12/15/2022]
Abstract
Angiogenesis and immunosuppression work hand-in-hand in the renal cell carcinoma (RCC) microenvironment. Tumor growth is associated with impaired antitumor immune response in RCC, which involves T cells, natural killer cells, dendritic cells (DCs) and macrophages. Vascular endothelial growth factor receptor (VEGFR), such as sorafenib, sunitinib, pazopanib and axitinib, and mammalian target of rapamycin (mTOR) inhibitors, such as temsirolimus and everolimus, do exert both antiangiogenic and immunomodulatory functions. Indeed, these agents affect neutrophil migration, as well as T lymphocyte-DC cross-talk, DC maturation and immune cell metabolism and reactivity. In this review, we overview the essential role of innate and adaptive immune response in RCC proliferation, invasion and metastasis and the relationship between tumor-associated immune cells and the response to targeted agents approved for the treatment of metastatic RCC.
Collapse
Affiliation(s)
- Matteo Santoni
- Medical Oncology AOU Ospedali Riuniti, Polytechnic University of the Marche Region, Ancona, Italy
| | | | | | | | | | | | | |
Collapse
|
11
|
Jaini R, Rayman P, Cohen PA, Finke JH, Tuohy VK. Combination of sunitinib with anti-tumor vaccination inhibits T cell priming and requires careful scheduling to achieve productive immunotherapy. Int J Cancer 2013; 134:1695-705. [PMID: 24105638 DOI: 10.1002/ijc.28488] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Revised: 07/30/2013] [Accepted: 08/29/2013] [Indexed: 02/02/2023]
Abstract
Sunitinib, a protein tyrosine kinase inhibitor is the frontline therapy for renal and gastrointestinal cancers. We hypothesized that by virtue of its well documented tumor apoptosis and immune adjuvant properties, combination of Sunitinib with anti-tumor immunotherapeutics will provide synergistic inhibition of tumor growth. Our study was designed to evaluate the impact of Sunitinib on immunotherapy mediated anti-tumor immune responses and evaluate its efficacy as a combinatorial therapy with tumor targeted immunotherapeutic vaccination. Mice immunized with recombinant α-lactalbumin, a lactation protein expressed on majority of breast tumors were treated with 1 mg of Sunitinib for seven consecutive days beginning (1) concurrently, on the day of α-lactalbumin immunization or (2) sequentially, on day 9 after immunization. Ten-day lymph nodes or 21 day spleens were tested by ELISPOT assays and flow cytometry to evaluate responsiveness to α-lactalbumin immunization in presence of Sunitinib and distribution of cells involved in T cell antigen priming and proliferation in different lymphoid compartments. In addition, therapeutic efficacy of the α-lactalbumin/ Sunitinib combination was evaluated by monitoring tumor growth in the 4T1 transplanted tumor model. Our studies reveal that concurrent administration of Sunitinib with active vaccination against a targeted tumor antigen inhibits priming to the immunogen due to a drastic decrease in CD11b+CD11c+ antigen presenting cells, leading to failure of vaccination. However, sequential delivery of Sunitinib timed to avoid the priming phase of vaccination results in the desired vaccination mediated boost in immune responses.
Collapse
Affiliation(s)
- Ritika Jaini
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | | | | | | | | |
Collapse
|
12
|
Krieg S, Ullrich E. Novel immune modulators used in hematology: impact on NK cells. Front Immunol 2013; 3:388. [PMID: 23316191 PMCID: PMC3539673 DOI: 10.3389/fimmu.2012.00388] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2012] [Accepted: 12/04/2012] [Indexed: 01/13/2023] Open
Abstract
There is a wide range of important pharmaceuticals used in treatment of cancer. Besides their known effects on tumor cells, there is growing evidence for modulation of the immune system. Immunomodulatory drugs (IMiDs®) play an important role in the treatment of patients with multiple myeloma or myelodysplastic syndrome and have already demonstrated antitumor, anti-angiogenic, and immunostimulating effects, in particular on natural killer (NK) cells. Tyrosine kinase inhibitors are directly targeting different kinases and are known to regulate effector NK cells and expression of NKG2D ligands (NKG2DLs) on tumor cells. Demethylating agents, histone deacetylases, and proteasome inhibitors interfere with the epigenetic regulation and protein degradation of malignant cells. There are first hints that these drugs also sensitize tumor cells to chemotherapy, radiation, and NK cell-mediated cytotoxicity by enhanced expression of TRAIL and NKG2DLs. However, these pharmaceuticals may also impair NK cell function in a dose- and time-dependent manner. In summary, this review provides an update on the effects of different novel molecules on the immune system focusing NK cells.
Collapse
Affiliation(s)
- Stephanie Krieg
- Hematology and Oncology, Department of Internal Medicine 5, University of Erlangen-Nuremberg Erlangen, Germany
| | | |
Collapse
|
13
|
Modulation of immunity by antiangiogenic molecules in cancer. Clin Dev Immunol 2012; 2012:492920. [PMID: 23320019 PMCID: PMC3540780 DOI: 10.1155/2012/492920] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Accepted: 12/10/2012] [Indexed: 12/18/2022]
Abstract
In the last decades a new class of therapeutic drugs have been developed that block tumor angiogenesis. These antiangiogenic molecules, which target VEGF or VEGFR, PDGFR, and c-kit, can act not only on endothelial cells but also on immune cells. Some antiangiogenic molecules inhibit the development of immunosuppressive mechanisms developed by the tumors to escape the immune system (such as regulatory T cells, myeloid-derived suppressor cells, and immunosuppressive cytokines). These immunomodulatory effects must be characterized in detail to enable a better prescription of these treatments. In this paper we will focus on the impact of anti-angiogenic drugs on immunosuppression and their potential combination with immunotherapeutic strategies. Interestingly, immune parameters or their modulation during treatment could serve as potential biomarkers of response or resistance to anti-angiogenic therapies.
Collapse
|