1
|
Jiang QY, Xue RY. Neutrophil-to-lymphocyte ratio and platelet-to-lymphocyte ratio: Markers predicting immune-checkpoint inhibitor efficacy and immune-related adverse events. World J Gastrointest Oncol 2024; 16:577-582. [PMID: 38577447 PMCID: PMC10989358 DOI: 10.4251/wjgo.v16.i3.577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/14/2023] [Accepted: 01/18/2024] [Indexed: 03/12/2024] Open
Abstract
We conducted a comprehensive review of existing prediction models pertaining to the efficacy of immune-checkpoint inhibitor (ICI) and the occurrence of immune-related adverse events (irAEs). The predictive potential of neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR) in determining ICI effectiveness has been extensively investigated, while limited research has been conducted on predicting irAEs. Furthermore, the combined model incorporating NLR and PLR, either with each other or in conjunction with additional markers such as carcinoembryonic antigen, exhibits superior predictive capabilities compared to individual markers alone. NLR and PLR are promising markers for clinical applications. Forthcoming models ought to incorporate established efficacious models and newly identified ones, thereby constituting a multifactor composite model. Furthermore, efforts should be made to explore effective clinical application approaches that enhance the predictive accuracy and efficiency.
Collapse
Affiliation(s)
- Qiu-Yu Jiang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Shanghai Institute of Liver Disease, Fudan University, Shanghai 200032, China
| | - Ru-Yi Xue
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Shanghai Institute of Liver Disease, Fudan University, Shanghai 200032, China
- Department of Gastroenterology and Hepatology, Shanghai Baoshan District Wusong Central Hospital (Zhongshan Hospital Wusong Branch, Fudan University), Shanghai 200940, China
| |
Collapse
|
2
|
de Joode K, Heersche N, Basak EA, Bins S, van der Veldt AAM, van Schaik RHN, Mathijssen RHJ. Review - The impact of pharmacogenetics on the outcome of immune checkpoint inhibitors. Cancer Treat Rev 2024; 122:102662. [PMID: 38043396 DOI: 10.1016/j.ctrv.2023.102662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/22/2023] [Accepted: 11/23/2023] [Indexed: 12/05/2023]
Abstract
The development of immune checkpoint inhibitors (ICIs) has a tremendous effect on the treatment options for multiple types of cancer. Nonetheless, there is a large interpatient variability in response, survival, and the development of immune-related adverse events (irAEs). Pharmacogenetics is the general term for germline genetic variations, which may cause the observed interindividual differences in response or toxicity to treatment. These genetic variations can either be single-nucleotide polymorphisms (SNPs) or structural variants, such as gene deletions, amplifications or rearrangements. For ICIs, pharmacogenetic variation in the human leukocyte antigen molecules has also been studied with regard to treatment outcome. This review presents a summary of the literature regarding the pharmacogenetics of ICI treatment, discusses the most important known genetic variations and offers recommendations on the application of pharmacogenetics for ICI treatment.
Collapse
Affiliation(s)
- Karlijn de Joode
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Niels Heersche
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands; Department of Clinical Chemistry, Erasmus MC, Erasmus University Hospital, Rotterdam, the Netherlands
| | - Edwin A Basak
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Sander Bins
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Astrid A M van der Veldt
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands; Department of Radiology & Nuclear Medicine, Erasmus MC, Erasmus University Hospital, Rotterdam, the Netherlands
| | - Ron H N van Schaik
- Department of Clinical Chemistry, Erasmus MC, Erasmus University Hospital, Rotterdam, the Netherlands
| | - Ron H J Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands.
| |
Collapse
|
3
|
Boutros A, Carosio R, Campanella D, Spagnolo F, Banelli B, Morabito A, Pistillo M, Croce E, Cecchi F, Pronzato P, Queirolo P, Raposio E, Fontana V, Tanda E. The predictive and prognostic role of single nucleotide gene variants of PD-1 and PD-L1 in patients with advanced melanoma treated with PD-1 inhibitors. IMMUNO-ONCOLOGY TECHNOLOGY 2023; 20:100408. [PMID: 38192613 PMCID: PMC10772261 DOI: 10.1016/j.iotech.2023.100408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/10/2024]
Abstract
Background Despite having revolutionized the treatment paradigm for advanced melanoma, not all patients benefit from immune checkpoint inhibitor therapy. To date, there are no predictive biomarkers for response or the occurrence of immune-related adverse events (irAEs) to programmed cell death protein 1 (PD-1) inhibitors. Our aim was to investigate the predictive and prognostic role of single nucleotide variants (SNVs) of genes involved in the PD-1 axis. Methods We analysed, in metastatic melanoma patients treated with nivolumab or pembrolizumab, five PD-1 SNVs, namely PD1.3 G>A (rs11568821), PD1.5 C>T (rs2227981), PD1.6 G>A (rs10204525), PD1.7 T>C(rs7421861), PD1.10 C>G (rs5582977) and three programmed death-ligand 1 (PD-L1) SNVs: +8293 C>A (rs2890658), PD-L1 C>T (rs2297136) and PD-L1 G>C (rs4143815). Association of SNV genotypic frequencies with best overall response to PD-1 inhibitors and development of irAEs were estimated through a modified Poisson regression. A Cox regression modelling approach was applied to evaluate the SNV association with OS. Results A total of 125 patients with advanced melanoma were included in the analysis. A reduction in irAEs risk was observed in patients carrying the PD-L1 +8293 C/A genotype compared with those carrying the C/C genotype (risk ratio = 0.45; 95% CL 0.22-0.93; P = 0.031). A trend for a reduction in irAEs was also observed with the PD1.5 T allele (risk ratio = 0.70, 95% confidence limits 0.48-1.01 versus C allele). None of the SNVs was associated with response to therapy. Finally, a survival benefit was observed in patients harbouring the PD1.7 C/C genotype (hazard ratio = 0.37; 95% confidence limits 0.14-0.96; P = 0.028) in the homozygous model. Conclusions Our study showed that PD-1.5 and PD-L1 +8293 SNVs may play a role as a predictive biomarker of development of irAEs to PD-1 inhibitors. PD1.7 SNV may also be associated with a reduction of the risk of death, although further translational research is needed to confirm these results.
Collapse
Affiliation(s)
- A. Boutros
- Skin Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, Genoa
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genoa
| | - R. Carosio
- Tumor Epigenetics Unit, IRCCS Ospedale Policlinico San Martino, Genoa
| | - D. Campanella
- Clinical Epidemiology Unit, IRCCS Ospedale Policlinico San Martino, Genoa
| | - F. Spagnolo
- Skin Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, Genoa
- Department of Surgical Sciences and Integrated Diagnostics (DISC), Plastic Surgery Division, University of Genova, Genoa
| | - B. Banelli
- Tumor Epigenetics Unit, IRCCS Ospedale Policlinico San Martino, Genoa
| | - A. Morabito
- Tumor Epigenetics Unit, IRCCS Ospedale Policlinico San Martino, Genoa
| | - M.P. Pistillo
- Tumor Epigenetics Unit, IRCCS Ospedale Policlinico San Martino, Genoa
| | - E. Croce
- Skin Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, Genoa
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genoa
| | - F. Cecchi
- Skin Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, Genoa
| | - P. Pronzato
- Skin Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, Genoa
| | - P. Queirolo
- Division of Melanoma Sarcoma and Rare Tumors, IRCCS European Institute of Oncology, Milan, Italy
| | - E. Raposio
- Department of Surgical Sciences and Integrated Diagnostics (DISC), Plastic Surgery Division, University of Genova, Genoa
| | - V. Fontana
- Clinical Epidemiology Unit, IRCCS Ospedale Policlinico San Martino, Genoa
| | - E.T. Tanda
- Skin Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, Genoa
| |
Collapse
|
4
|
Menotti S, Giampietro A, Raia S, Veleno M, Angelini F, Tartaglione T, Gaudino S, Doglietto F, De Marinis L, Pontecorvi A, Bianchi A, Chiloiro S. Unveiling the Etiopathogenic Spectrum of Hypophysitis: A Narrative Review. J Pers Med 2023; 13:1210. [PMID: 37623461 PMCID: PMC10455260 DOI: 10.3390/jpm13081210] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 07/25/2023] [Accepted: 07/27/2023] [Indexed: 08/26/2023] Open
Abstract
Hypophysitis, a rare inflammatory disorder of the pituitary gland, has seen an uptick in reported cases in recent years. Our objective is to summarize the most recent research on the etiopathogenesis, molecular mechanisms, and genetics of both primary and secondary hypophysitis. Primary autoimmune hypophysitis (PAH): During the acute phase of the disease, the pituitary gland in enlarged due to the infiltration of T and B lymphocytes. The chronic phase is characterized by progressive and irreversible pituitary atrophy. APA may play a role in the management, diagnosis, and prognosis of PAH. Specific autoantibodies such as anti-GH, anti-PIT-1, and anti-T-PIT have been found in patients with hypophysitis and hypopituitarism. A recent study suggested that a mechanism of escaping clonal deletion and mounting an immune response against self antigens can explain the unusual nature of the immune response observed in PAH patients. A cytokine array shows the presence of gamma-interferon and interleukin-17. Patients carrying mutations in the PIT1 or PROP1 genes may present PAH. Individuals carrying the HLA DQ8 haplotype are four times more likely to develop PAH. Immune checkpoint inhibitors induce hypophysitis (IIHs): IIHs is an increasingly frequent toxicity of in patients on treatment with inhibitors targeting cytotoxic T-lymphocyte antigen 4 (CTLA-4) and programmed cell death-1 (PD-1). ICIs inhibit the CTLA-4 pathway, leading to overactivation of T lymphocytes. The binding of PD-1/PD-L1 suppresses the activity of T cells, promotes the conversion of T-helpers into T-regulatory cells, and activates pro-survival signaling pathways in cancer cells. Cytokines play a crucial role in IIHs. B-cell infiltration has been observed in IIHs, suggesting that antibody-mediated pituitary injury may contribute. Genetic polymorphisms of CTLA-4 and PD-1 genes can increase the risk of IIHs. HLA alleles may also be involved in the onset of IIHs; this HLA association presents a possible alternative mechanistic hypothesis. IIHs may also be linked to a paraneoplastic syndrome triggered by ectopic expression of pituitary specific antigens. SARS-CoV-2-related hypophysitis: Recently, the literature has reported occurrences of hypophysitis associated with the SARS-CoV-2 virus; long COVID-19 may also present as infundibulo-neuro-hypophysitis. The virus enters the central nervous system because of its distinct interaction with angiotensin-converting enzyme receptors via spike proteins binding the capillary endothelium, and it directly damages the pituitary cells. The effect of SARS-CoV-2 can occur indirectly through inflammation and the release of cytokines. The exact mechanism remains ambiguous. The available data on endocrine complications associated with the SARS-CoV-2 vaccine are scant. Nonetheless, isolated cases of hypophysitis have been documented. Treatment of hypophysitis: Glucocorticoids are the cornerstone in managing primary hypophysitis, given their targeted action on inflammation. A better understanding of the etiopathogenesis and molecular mechanism of hypophysitis can lead to more effective and personalized treatment strategies.
Collapse
Affiliation(s)
- Sara Menotti
- Pituitary Unit, Department of Endocrinology and Metabolism, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (S.M.); (A.G.); (S.R.); (M.V.); (F.A.); (L.D.M.); (A.P.); (S.C.)
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (T.T.); (S.G.); (F.D.)
| | - Antonella Giampietro
- Pituitary Unit, Department of Endocrinology and Metabolism, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (S.M.); (A.G.); (S.R.); (M.V.); (F.A.); (L.D.M.); (A.P.); (S.C.)
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (T.T.); (S.G.); (F.D.)
| | - Salvatore Raia
- Pituitary Unit, Department of Endocrinology and Metabolism, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (S.M.); (A.G.); (S.R.); (M.V.); (F.A.); (L.D.M.); (A.P.); (S.C.)
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (T.T.); (S.G.); (F.D.)
| | - Miriam Veleno
- Pituitary Unit, Department of Endocrinology and Metabolism, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (S.M.); (A.G.); (S.R.); (M.V.); (F.A.); (L.D.M.); (A.P.); (S.C.)
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (T.T.); (S.G.); (F.D.)
| | - Flavia Angelini
- Pituitary Unit, Department of Endocrinology and Metabolism, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (S.M.); (A.G.); (S.R.); (M.V.); (F.A.); (L.D.M.); (A.P.); (S.C.)
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (T.T.); (S.G.); (F.D.)
| | - Tommaso Tartaglione
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (T.T.); (S.G.); (F.D.)
- Department of Radiodiagnostic, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Simona Gaudino
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (T.T.); (S.G.); (F.D.)
- Department of Radiodiagnostic, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Francesco Doglietto
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (T.T.); (S.G.); (F.D.)
- Department of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Laura De Marinis
- Pituitary Unit, Department of Endocrinology and Metabolism, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (S.M.); (A.G.); (S.R.); (M.V.); (F.A.); (L.D.M.); (A.P.); (S.C.)
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (T.T.); (S.G.); (F.D.)
| | - Alfredo Pontecorvi
- Pituitary Unit, Department of Endocrinology and Metabolism, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (S.M.); (A.G.); (S.R.); (M.V.); (F.A.); (L.D.M.); (A.P.); (S.C.)
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (T.T.); (S.G.); (F.D.)
| | - Antonio Bianchi
- Pituitary Unit, Department of Endocrinology and Metabolism, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (S.M.); (A.G.); (S.R.); (M.V.); (F.A.); (L.D.M.); (A.P.); (S.C.)
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (T.T.); (S.G.); (F.D.)
| | - Sabrina Chiloiro
- Pituitary Unit, Department of Endocrinology and Metabolism, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (S.M.); (A.G.); (S.R.); (M.V.); (F.A.); (L.D.M.); (A.P.); (S.C.)
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (T.T.); (S.G.); (F.D.)
| |
Collapse
|
5
|
Papachristos A, Patel J, Vasileiou M, Patrinos GP. Dose Optimization in Oncology Drug Development: The Emerging Role of Pharmacogenomics, Pharmacokinetics, and Pharmacodynamics. Cancers (Basel) 2023; 15:3233. [PMID: 37370844 DOI: 10.3390/cancers15123233] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Drugs' safety and effectiveness are evaluated in randomized, dose-ranging trials in most therapeutic areas. However, this is only sometimes feasible in oncology, and dose-ranging studies are mainly limited to Phase 1 clinical trials. Moreover, although new treatment modalities (e.g., small molecule targeted therapies, biologics, and antibody-drug conjugates) present different characteristics compared to cytotoxic agents (e.g., target saturation limits, wider therapeutic index, fewer off-target side effects), in most cases, the design of Phase 1 studies and the dose selection is still based on the Maximum Tolerated Dose (MTD) approach used for the development of cytotoxic agents. Therefore, the dose was not optimized in some cases and was modified post-marketing (e.g., ceritinib, dasatinib, niraparib, ponatinib, cabazitaxel, and gemtuzumab-ozogamicin). The FDA recognized the drawbacks of this approach and, in 2021, launched Project Optimus, which provides the framework and guidance for dose optimization during the clinical development stages of anticancer agents. Since dose optimization is crucial in clinical development, especially of targeted therapies, it is necessary to identify the role of pharmacological tools such as pharmacogenomics, therapeutic drug monitoring, and pharmacodynamics, which could be integrated into all phases of drug development and support dose optimization, as well as the chances of positive clinical outcomes.
Collapse
Affiliation(s)
| | - Jai Patel
- Department of Cancer Pharmacology and Pharmacogenomics, Levine Cancer Institute, Atrium Health, Charlotte, NC 28204, USA
| | - Maria Vasileiou
- Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, 16121 Athens, Greece
| | - George P Patrinos
- Laboratory of Pharmacogenomics and Individualized Therapy, Department of Pharmacy, School of Health Sciences, University of Patras, 26504 Patras, Greece
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| |
Collapse
|
6
|
Les I, Martínez M, Pérez-Francisco I, Cabero M, Teijeira L, Arrazubi V, Torrego N, Campillo-Calatayud A, Elejalde I, Kochan G, Escors D. Predictive Biomarkers for Checkpoint Inhibitor Immune-Related Adverse Events. Cancers (Basel) 2023; 15:1629. [PMID: 36900420 PMCID: PMC10000735 DOI: 10.3390/cancers15051629] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/24/2023] [Accepted: 02/28/2023] [Indexed: 03/09/2023] Open
Abstract
Immune-checkpoint inhibitors (ICIs) are antagonists of inhibitory receptors in the immune system, such as the cytotoxic T-lymphocyte-associated antigen-4, the programmed cell death protein-1 and its ligand PD-L1, and they are increasingly used in cancer treatment. By blocking certain suppressive pathways, ICIs promote T-cell activation and antitumor activity but may induce so-called immune-related adverse events (irAEs), which mimic traditional autoimmune disorders. With the approval of more ICIs, irAE prediction has become a key factor in improving patient survival and quality of life. Several biomarkers have been described as potential irAE predictors, some of them are already available for clinical use and others are under development; examples include circulating blood cell counts and ratios, T-cell expansion and diversification, cytokines, autoantibodies and autoantigens, serum and other biological fluid proteins, human leucocyte antigen genotypes, genetic variations and gene profiles, microRNAs, and the gastrointestinal microbiome. Nevertheless, it is difficult to generalize the application of irAE biomarkers based on the current evidence because most studies have been retrospective, time-limited and restricted to a specific type of cancer, irAE or ICI. Long-term prospective cohorts and real-life studies are needed to assess the predictive capacity of different potential irAE biomarkers, regardless of the ICI type, organ involved or cancer site.
Collapse
Affiliation(s)
- Iñigo Les
- Internal Medicine Department, Navarre University Hospital, 31008 Pamplona, Spain
- Autoimmune Diseases Unit, Internal Medicine Department, Navarre University Hospital, 31008 Pamplona, Spain
- Inflammatory and Immune-Mediated Diseases Group, Instituto de Investigación Sanitaria de Navarra (IdISNA), Navarrabiomed-Public University of Navarre, 31008 Pamplona, Spain
| | - Mireia Martínez
- Osakidetza Basque Health Service, Department of Medical Oncology, Araba University Hospital, 01009 Vitoria-Gasteiz, Spain
- Lung Cancer Research Group, Bioaraba Health Research Institute, 01006 Vitoria-Gasteiz, Spain
| | - Inés Pérez-Francisco
- Breast Cancer Research Group, Bioaraba Health Research Institute, 01006 Vitoria-Gasteiz, Spain
| | - María Cabero
- Clinical Trials Platform, Bioaraba Health Research Institute, 01006 Vitoria-Gasteiz, Spain
| | - Lucía Teijeira
- Medical Oncology Department, Navarre University Hospital, 31008 Pamplona, Spain
| | - Virginia Arrazubi
- Medical Oncology Department, Navarre University Hospital, 31008 Pamplona, Spain
| | - Nuria Torrego
- Osakidetza Basque Health Service, Department of Medical Oncology, Araba University Hospital, 01009 Vitoria-Gasteiz, Spain
- Lung Cancer Research Group, Bioaraba Health Research Institute, 01006 Vitoria-Gasteiz, Spain
| | - Ana Campillo-Calatayud
- Inflammatory and Immune-Mediated Diseases Group, Instituto de Investigación Sanitaria de Navarra (IdISNA), Navarrabiomed-Public University of Navarre, 31008 Pamplona, Spain
| | - Iñaki Elejalde
- Internal Medicine Department, Navarre University Hospital, 31008 Pamplona, Spain
- Autoimmune Diseases Unit, Internal Medicine Department, Navarre University Hospital, 31008 Pamplona, Spain
- Inflammatory and Immune-Mediated Diseases Group, Instituto de Investigación Sanitaria de Navarra (IdISNA), Navarrabiomed-Public University of Navarre, 31008 Pamplona, Spain
| | - Grazyna Kochan
- Oncoimmunology Group, Instituto de Investigación Sanitaria de Navarra (IdISNA), Navarrabiomed-Public University of Navarre, 31008 Pamplona, Spain
| | - David Escors
- Oncoimmunology Group, Instituto de Investigación Sanitaria de Navarra (IdISNA), Navarrabiomed-Public University of Navarre, 31008 Pamplona, Spain
| |
Collapse
|
7
|
Zheng LP, Yang J, Chen XW, Li LC, Sun JG. Correlation of preclinical and clinical biomarkers with efficacy and toxicity of cancer immunotherapy. Ther Adv Med Oncol 2023; 15:17588359231163807. [PMID: 37113734 PMCID: PMC10126660 DOI: 10.1177/17588359231163807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 02/27/2023] [Indexed: 04/29/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revealed significant clinical values in different solid tumors and hematological malignancy, changing the landscape for the treatment of multiple types of cancer. However, only a subpopulation of patients has obvious tumor response and long-term survival after ICIs treatment, and many patients may experience other undesirable clinical features. Therefore, biomarkers are critical for patients to choose exact optimum therapy. Here, we reviewed existing preclinical and clinical biomarkers of immunotherapeutic efficacy and immune-related adverse events (irAEs). Based on efficacy prediction, pseudoprogression, hyperprogressive disease, or irAEs, these biomarkers were divided into cancer cell-derived biomarkers, tumor microenvironment-derived biomarkers, host-derived biomarkers, peripheral blood biomarkers, and multi-modal model and artificial intelligence assessment-based biomarkers. Furthermore, we describe the relation between ICIs efficacy and irAEs. This review provides the overall perspective of biomarkers of immunotherapeutic outcome and irAEs prediction during ICIs treatment.
Collapse
Affiliation(s)
| | | | - Xie-Wan Chen
- Department of Basic Medicine, Army Medical University, Chongqing, China
| | - Ling-Chen Li
- Cancer Institute, Xinqiao Hospital, Army Medical University, Chongqing, China
| | | |
Collapse
|
8
|
Liu W, Luo Z, Liu Y, Sun B. Current landscape and tailored management of immune-related adverse events. Front Pharmacol 2023; 14:1078338. [PMID: 36950013 PMCID: PMC10025325 DOI: 10.3389/fphar.2023.1078338] [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: 10/24/2022] [Accepted: 02/24/2023] [Indexed: 03/08/2023] Open
Abstract
Unprecedented advances have been made in immune checkpoint inhibitors (ICIs) in the treatment of cancer. However, the overall benefits from ICIs are impaired by the increasing incidence of immune-related adverse events (irAEs). Although several factors and mechanisms have been proposed in the development of irAEs, there is still incomprehensive understanding of irAEs. Therefore, it is urgent to identify certain risk factors and biomarkers that predict the development of irAEs, as well as to understand the underlying mechanisms of these adverse events. Herein, we comprehensively summarize the state-of-the-art knowledge about clinical features and the related risk factors of irAEs. Particularly, we also discuss relevant mechanisms of irAEs and address the mechanism-based strategies, aiming to develop a tailored management approach for irAEs.
Collapse
Affiliation(s)
- Wenhui Liu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Zhiying Luo
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Yiping Liu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Bao Sun
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Bao Sun,
| |
Collapse
|
9
|
Germline genetic variation and predicting immune checkpoint inhibitor induced toxicity. NPJ Genom Med 2022; 7:73. [PMID: 36564402 PMCID: PMC9789157 DOI: 10.1038/s41525-022-00345-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 12/07/2022] [Indexed: 12/25/2022] Open
Abstract
Immune checkpoint inhibitor (ICI) therapy has revolutionised the treatment of various cancer types. ICIs reinstate T-cell function to elicit an anti-cancer immune response. The resulting immune response can however have off-target effects which manifest as autoimmune type serious immune-related adverse events (irAE) in ~10-55% of patients treated. It is currently challenging to predict both who will experience irAEs and to what severity. Identification of patients at high risk of serious irAE would revolutionise patient care. While the pathogenesis driving irAE development is still unclear, host genetic factors are proposed to be key determinants of these events. This review presents current evidence supporting the role of the host genome in determining risk of irAE. We summarise the spectrum and timing of irAEs following treatment with ICIs and describe currently reported germline genetic variation associated with expression of immuno-modulatory factors within the cancer immunity cycle, development of autoimmune disease and irAE occurrence. We propose that germline genetic determinants of host immune function and autoimmune diseases could also explain risk of irAE development. We also endorse genome-wide association studies of patients being treated with ICIs to identify genetic variants that can be used in polygenic risk scores to predict risk of irAE.
Collapse
|
10
|
Chiloiro S, Bianchi A, Giampietro A, Milardi D, De Marinis L, Pontecorvi A. The changing clinical spectrum of endocrine adverse events in cancer immunotherapy. Trends Endocrinol Metab 2022; 33:87-104. [PMID: 34895977 DOI: 10.1016/j.tem.2021.10.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 10/18/2021] [Accepted: 10/29/2021] [Indexed: 12/15/2022]
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of several malignancies, improving patient survival and quality of life. Endocrinopathies have emerged as a clinically significant group of immune-related adverse events (IRAEs). Although the mechanism of ICI toxicities has not been clarified, inhibition of immune checkpoints reduces immune tolerance to autoantigens, resulting in the development of autoimmunity disorders. We report current evidence regarding endocrine IRAEs that may have diagnostic and therapeutic implications. Management should be focused on a multidisciplinary approach to reach a prompt diagnosis and an appropriate and safe treatment.
Collapse
Affiliation(s)
- Sabrina Chiloiro
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Roma, Italy; Unità Operativa Complessa (UOC) Endocrinology and Diabetology, Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Roma, Italy.
| | - Antonio Bianchi
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Roma, Italy; Unità Operativa Complessa (UOC) Endocrinology and Diabetology, Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Roma, Italy
| | - Antonella Giampietro
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Roma, Italy; Unità Operativa Complessa (UOC) Endocrinology and Diabetology, Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Roma, Italy
| | - Domenico Milardi
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Roma, Italy; Unità Operativa Complessa (UOC) Endocrinology and Diabetology, Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Roma, Italy
| | - Laura De Marinis
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Roma, Italy; Unità Operativa Complessa (UOC) Endocrinology and Diabetology, Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Roma, Italy
| | - Alfredo Pontecorvi
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Roma, Italy; Unità Operativa Complessa (UOC) Endocrinology and Diabetology, Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Roma, Italy
| |
Collapse
|
11
|
Smithy JW, Faleck DM, Postow MA. Facts and Hopes in Prediction, Diagnosis, and Treatment of Immune-Related Adverse Events. Clin Cancer Res 2021; 28:1250-1257. [PMID: 34921018 DOI: 10.1158/1078-0432.ccr-21-1240] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/20/2021] [Accepted: 12/07/2021] [Indexed: 11/16/2022]
Abstract
Over the past decade, the use of immune checkpoint inhibitors (ICI) has expanded across a wide spectrum of oncology indications. Immune-related adverse events (irAE) from ICIs represent a significant source of morbidity, and in rare instances, can lead to treatment-related mortality. There are significant opportunities to better identify patients at increased risk for immune-related toxicity, diagnose irAEs more accurately and earlier in their course, and develop more individualized therapeutic strategies once complications arise. Clinical characteristics, germline and somatic genetic features, microbiome composition, and circulating biomarkers have all been associated with higher risk of developing irAEs in retrospective series. Many of these data suggest that both antitumor and anti-host ICI-associated immune reactions may be driven by common features of either the tumor or the patient's preexisting immune milieu. While irAE diagnosis is currently based on clinical history, exclusion of alternative etiologies, and sometimes pathologic confirmation, novel blood-based and radiographic assays are in development to identify these complications more precisely. Anecdotal reports and small case series have highlighted the potential role of targeted immunomodulatory agents to treat irAEs, though further prospective investigation is needed to evaluate more rigorously their use in these settings. In this review, we highlight the current state of knowledge about predicting, diagnosing, and treating irAEs with a translational focus and discuss emerging strategies which aim to improve each of these domains.
Collapse
Affiliation(s)
- James W Smithy
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - David M Faleck
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Weill Cornell Medical College, New York, New York
| | - Michael A Postow
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York. .,Weill Cornell Medical College, New York, New York
| |
Collapse
|
12
|
Chiloiro S, Russo F, Tartaglione T, Capoluongo ED. Molecular and Genetic Immune Biomarkers of Primary and Immune-Therapy Induced Hypophysitis: From Laboratories to the Clinical Practice. J Pers Med 2021; 11:1026. [PMID: 34683167 PMCID: PMC8537090 DOI: 10.3390/jpm11101026] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/15/2021] [Accepted: 10/12/2021] [Indexed: 12/12/2022] Open
Abstract
Hypophysitis is a rare and potentially life-threatening disease, characterized by an elevated risk of complications, such as the occurrence of acute central hypoadrenalism, persistent hypopituitarism, or the extension of the inflammatory process to the neighboring neurological structures. In recent years, a large number of cases has been described. The diagnosis of hypophysitis is complex because it is based on clinical and radiological criteria. Due to this, the integration of molecular and genetic biomarkers can help physicians in the diagnosis of hypophysitis and play a role in predicting disease outcome. In this paper, we review current knowledge about molecular and genetic biomarkers of hypophysitis with the aim of suggesting a possible integration of these biomarkers in clinical practice.
Collapse
Affiliation(s)
- Sabrina Chiloiro
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
- UOC Endocrinology and Diabetology, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy
| | - Filippo Russo
- Department of Molecular Medicine and Medical Biotechnology, Federico II University-CEINGE, 80126 Naples, Italy;
| | - Tommaso Tartaglione
- Department of Radiological and Haematological Sciences, Università Cattolica del Sacro Cuore, 00168 Roma, Italy;
- Department of Radiology and Diagnostic Imaging, Istituto Dermopatico dell’Immacolata, IDI-IRCCS, 00167 Roma, Italy
| | | |
Collapse
|
13
|
Abdel-Wahab N, Diab A, Yu RK, Futreal A, Criswell LA, Tayar JH, Dadu R, Shannon V, Shete SS, Suarez-Almazor ME. Genetic determinants of immune-related adverse events in patients with melanoma receiving immune checkpoint inhibitors. Cancer Immunol Immunother 2021; 70:1939-1949. [PMID: 33409738 PMCID: PMC10992432 DOI: 10.1007/s00262-020-02797-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 11/08/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) can cause profound immune-related adverse events (irAEs). The host genetic background is likely to play a role in irAE susceptibility because the presentation of toxicity varies among patients and many do not develop irAEs despite continued ICI use. We sought to identify potential genetic markers conferring risk for irAEs. METHODS We conducted a pilot exploratory study in 89 melanoma patients who received ICIs (44 with irAEs, and 45 without irAEs after at least 1 year from starting treatment). Genotyping was performed using the Infinium Multi-Ethnic Global-8 v1.0 Bead Chip. The genotype data were extracted using PLINK (v1.90b3.34) and processed for quality control. Population structure-based clustering was carried out using IBS matrix, pairwise population concordance test (p < 1 × 10-3), and phenotype distribution for all study participants, resulting in seven population structure-based clusters. In the analytical stage, 599,931 variants in autosomal chromosomes were included for the association study. The association test was performed using an additive genetic model with exact logistic regression, adjusted for age, sex, and population cluster. RESULTS A total of 30 variants or single-nucleotide polymorphisms with p < 1 × 10-4 were identified; 12 were associated with an increased risk of irAEs, and the remaining 18 were associated with a decreased risk. Overall, nine of the identified single-nucleotide polymorphisms mapped to eight unique genes that have been associated with autoimmunity or inflammatory diseases. CONCLUSION Several genetic variants associated with irAEs were identified. Additional larger studies are needed to validate these findings and establish their potential functional relevance.
Collapse
Affiliation(s)
- Noha Abdel-Wahab
- Section of Rheumatology and Clinical Immunology, Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Rheumatology and Rehabilitation, Faculty of Medicine, Assiut University Hospitals, Assiut, Egypt
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Adi Diab
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Robert K Yu
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Andrew Futreal
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lindsey A Criswell
- Russell/Engleman Rheumatology Research Center, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Jean H Tayar
- Section of Rheumatology and Clinical Immunology, Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ramona Dadu
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vickie Shannon
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sanjay S Shete
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Maria E Suarez-Almazor
- Section of Rheumatology and Clinical Immunology, Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Health Services Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
14
|
Zheng Y, Deng Z, Tang M, Xiao D, Cai P. Impact of genetic factors on platinum-induced gastrointestinal toxicity. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2020; 786:108324. [PMID: 33339576 DOI: 10.1016/j.mrrev.2020.108324] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 07/09/2020] [Accepted: 07/14/2020] [Indexed: 01/20/2023]
Abstract
Severe gastrointestinal (GI) toxicity is a common side effect after platinum-based chemotherapy. The incidence and severity of GI toxicity vary among patients with the same chemotherapy. Genetic factors involved in platinum transport, metabolism, detoxification, DNA repair, cell cycle control, and apoptosis pathways may account for the interindividual difference in GI toxicity. The influence of gene polymorphisms in the platinum pathway on GI toxicity has been extensively analyzed. Variations in study sample size, ethnicity, design, treatment schedule, dosing, endpoint definition, and assessment of toxicity make it difficult to precisely interpret the results. Hence, we conducted a review to summarize the most recent pharmacogenomics studies of GI toxicity in platinum-based chemotherapy and identify the most promising avenues for further research.
Collapse
Affiliation(s)
- Yi Zheng
- Hunan Provincial Maternal and Child Health Care Hospital, Changsha, 410008, People's Republic of China
| | - Zheng Deng
- Hunan Institute for Tuberculosis Control, Changsha, 410008, People's Republic of China; Hunan Chest Hospital, Changsha, 410008, People's Republic of China
| | - Mimi Tang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
| | - Di Xiao
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
| | - Pei Cai
- Hunan Provincial Maternal and Child Health Care Hospital, Changsha, 410008, People's Republic of China.
| |
Collapse
|
15
|
Simões AR, Fernández-Rozadilla C, Maroñas O, Carracedo Á. The Road so Far in Colorectal Cancer Pharmacogenomics: Are We Closer to Individualised Treatment? J Pers Med 2020; 10:E237. [PMID: 33228198 PMCID: PMC7711884 DOI: 10.3390/jpm10040237] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 12/12/2022] Open
Abstract
In recent decades, survival rates in colorectal cancer have improved greatly due to pharmacological treatment. However, many patients end up developing adverse drug reactions that can be severe or even life threatening, and that affect their quality of life. These remain a limitation, as they may force dose reduction or treatment discontinuation, diminishing treatment efficacy. From candidate gene approaches to genome-wide analysis, pharmacogenomic knowledge has advanced greatly, yet there is still huge and unexploited potential in the use of novel technologies such as next-generation sequencing strategies. This review summarises the road of colorectal cancer pharmacogenomics so far, presents considerations and directions to be taken for further works and discusses the path towards implementation into clinical practice.
Collapse
Affiliation(s)
- Ana Rita Simões
- Grupo de Medicina Xenómica, Universidade de Santiago de Compostela (USC), 15706 Santiago de Compostela, Spain; (A.R.S.); (O.M.); (Á.C.)
- Instituto de Investigación Sanitaria de Santiago (IDIS), 15706 Santiago de Compostela, Spain
| | - Ceres Fernández-Rozadilla
- Grupo de Medicina Xenómica, Universidade de Santiago de Compostela (USC), 15706 Santiago de Compostela, Spain; (A.R.S.); (O.M.); (Á.C.)
- Instituto de Investigación Sanitaria de Santiago (IDIS), 15706 Santiago de Compostela, Spain
| | - Olalla Maroñas
- Grupo de Medicina Xenómica, Universidade de Santiago de Compostela (USC), 15706 Santiago de Compostela, Spain; (A.R.S.); (O.M.); (Á.C.)
| | - Ángel Carracedo
- Grupo de Medicina Xenómica, Universidade de Santiago de Compostela (USC), 15706 Santiago de Compostela, Spain; (A.R.S.); (O.M.); (Á.C.)
- Instituto de Investigación Sanitaria de Santiago (IDIS), 15706 Santiago de Compostela, Spain
- Fundación Pública Galega de Medicina Xenómica; SERGAS, 15706 Santiago de Compostela, Spain
- Consorcio Centro de Investigación Biomédica en Red de Enfermedades Raras—CIBERER, 28029 Madrid, Spain
| |
Collapse
|
16
|
Pistillo MP, Carosio R, Grillo F, Fontana V, Mastracci L, Morabito A, Banelli B, Tanda E, Cecchi F, Dozin B, Gualco M, Salvi S, Spagnolo F, Poggi A, Queirolo P. Phenotypic characterization of tumor CTLA-4 expression in melanoma tissues and its possible role in clinical response to Ipilimumab. Clin Immunol 2020; 215:108428. [PMID: 32344017 DOI: 10.1016/j.clim.2020.108428] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 02/21/2020] [Accepted: 04/17/2020] [Indexed: 01/13/2023]
Abstract
The expression of the immune checkpoint molecule CTLA-4 has been almost exclusively studied in the T cell lineage, but increasing evidence has shown its expression on tumors with implications for immunotherapy. To date, the degree of expression of CTLA-4 on tumor cells as a predictive biomarker of response to immune checkpoint inhibitors has not been studied. In this report, we analyzed this issue in melanoma patients treated with CTLA-4 inhibitor Ipilimumab (IPI). We show that the level of CTLA-4 expression on melanoma cells is higher than that on tumor infiltrating lymphocytes (TIL) and it is associated with clinical response to IPI therapy supporting the idea of its possible role as a predictive biomarker.
Collapse
Affiliation(s)
- Maria Pia Pistillo
- IRCCS Ospedale Policlinico San Martino, Tumor Epigenetics Unit, Genova, Italy.
| | - Roberta Carosio
- IRCCS Ospedale Policlinico San Martino, Tumor Epigenetics Unit, Genova, Italy
| | - Federica Grillo
- University of Genova, Department of Surgical Sciences and Integrated Diagnostics (DISC), Genova, Italy; IRCCS Ospedale Policlinico San Martino, Anatomic Pathology Unit, Genova, Italy
| | - Vincenzo Fontana
- IRCCS Ospedale Policlinico San Martino, Clinical Epidemiology Unit, Genova, Italy
| | - Luca Mastracci
- University of Genova, Department of Surgical Sciences and Integrated Diagnostics (DISC), Genova, Italy; IRCCS Ospedale Policlinico San Martino, Anatomic Pathology Unit, Genova, Italy
| | - Anna Morabito
- IRCCS Ospedale Policlinico San Martino, Tumor Epigenetics Unit, Genova, Italy
| | - Barbara Banelli
- IRCCS Ospedale Policlinico San Martino, Tumor Epigenetics Unit, Genova, Italy
| | - Enrica Tanda
- IRCCS Ospedale Policlinico San Martino, Skin Cancer Unit, Genova, Italy
| | - Federica Cecchi
- IRCCS Ospedale Policlinico San Martino, Skin Cancer Unit, Genova, Italy
| | - Beatrice Dozin
- IRCCS Ospedale Policlinico San Martino, Clinical Epidemiology Unit, Genova, Italy
| | - Marina Gualco
- Ospedale Villa Scassi, ASL3, Anatomic Pathology Unit, Genova, Italy
| | - Sandra Salvi
- IRCCS Ospedale Policlinico San Martino, Anatomic Pathology Unit, Genova, Italy
| | | | - Alessandro Poggi
- IRCCS Ospedale Policlinico San Martino, Molecular Oncology and Angiogenesis Unit, Genova, Italy
| | - Paola Queirolo
- IEO, Division of Medical Oncology for Melanoma, Sarcoma, and Rare Tumors, European Institute of Oncology IRCCS, Milan, Italy
| |
Collapse
|
17
|
Connolly C, Bambhania K, Naidoo J. Immune-Related Adverse Events: A Case-Based Approach. Front Oncol 2019; 9:530. [PMID: 31293970 PMCID: PMC6598598 DOI: 10.3389/fonc.2019.00530] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 05/31/2019] [Indexed: 12/23/2022] Open
Abstract
Immunotherapy has heralded the advent of a new era in oncology. Immune checkpoint inhibitors (ICIs) enhance anti-tumor immunity, thereby reinvigorating a patient's immune system to fight cancer. While therapy with this class of agents has resulted in improved clinical outcomes for patients with multiple tumor types, a broad spectrum of immune-related adverse events (irAEs) may affect any organ system, with variable clinical presentations. Prompt recognition and management of irAEs are associated with improved irAE outcomes, and represents an important new clinical challenge for practicing oncologists. Herein, we provide a comprehensive case-based review of the most common and clinically-important irAEs, focussing on epidemiology, clinical manifestations, and management. We also examine future strategies that may provide meaningful insights into the prevention and management of irAEs.
Collapse
Affiliation(s)
- Caoilfhionn Connolly
- Department of Internal Medicine, Johns Hopkins Hospital, Baltimore, MD, United States
| | - Kalindi Bambhania
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, United States
| | - Jarushka Naidoo
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, United States
- Department of Oncology, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Baltimore, MD, United States
| |
Collapse
|
18
|
Shek D, Read SA, Ahlenstiel G, Piatkov I. Pharmacogenetics of anticancer monoclonal antibodies. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2019; 2:69-81. [PMID: 35582142 PMCID: PMC9019180 DOI: 10.20517/cdr.2018.20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 01/09/2019] [Accepted: 02/19/2019] [Indexed: 12/16/2022]
Abstract
Pharmacogenetics is the study of therapeutic and adverse responses to drugs based on an individual’s genetic background. Monoclonal antibodies (mAbs) are a rapidly evolving field in cancer therapy, however a number of newly developed and highly effective mAbs (e.g., anti-CTLA-4 and anti-PD-1) possess pharmacogenomic profiles that remain largely undefined. Since the first chemotherapeutic mAb Rituximab was approved in 1997 by the US Food and Drug Administration for cancer treatment, a broad number of other mAbs have been successfully developed and implemented into oncological practice. Nowadays, mAbs are considered as one of the most promising new approaches for cancer treatment. The efficacy of mAb treatment can however be significantly affected by genetic background, where genes responsible for antibody presentation and metabolism, for example, can seriously affect patient outcome. This review will focus on current anticancer mAb treatments, patient genetics that shape their efficacy, and the molecular pathways that bridge the two.
Collapse
Affiliation(s)
- Dmitrii Shek
- Blacktown Clinical School, Western Sydney University, Blacktown, NSW 2148, Australia
| | - Scott A Read
- Blacktown Clinical School, Western Sydney University, Blacktown, NSW 2148, Australia.,Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW 2145, Australia
| | - Golo Ahlenstiel
- Blacktown Clinical School, Western Sydney University, Blacktown, NSW 2148, Australia.,Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW 2145, Australia.,Blacktown Hospital, Blacktown, NSW 2148, Australia
| | | |
Collapse
|
19
|
Cappelli LC, Dorak MT, Bettinotti MP, Bingham CO, Shah AA. Association of HLA-DRB1 shared epitope alleles and immune checkpoint inhibitor-induced inflammatory arthritis. Rheumatology (Oxford) 2019; 58:476-480. [PMID: 30508191 PMCID: PMC6821338 DOI: 10.1093/rheumatology/key358] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 09/30/2018] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE To evaluate the frequency of HLA class I and II alleles associated with traditional forms of inflammatory arthritis in patients with immune checkpoint inhibitor (ICI)-induced inflammatory arthritis as compared with population controls. METHODS High-resolution HLA typing was performed on 27 patients with ICI-induced inflammatory arthritis and 726 healthy controls. Genotyping at the shared epitope (SE) locus (HLA DRB1) was performed on 220 RA cases. Allele-positivity rates and frequency of having at least one SE allele were compared using Fisher's exact test between ICI-induced inflammatory arthritis and healthy controls. Frequency of having at least one SE allele was also compared between ICI-induced inflammatory arthritis and RA cases. RESULTS Twenty-six patients with ICI-induced inflammatory arthritis were of European descent, and one was African American. In those 26 patients, 16 (61.5%) had at least one SE allele, significantly different from healthy controls of European descent, in whom 299 (41.2%) had at least one SE allele (odds ratio 2.3, P = 0.04). The allele-positivity rate of DRB1*04: 05 was also higher in the ICI-induced inflammatory arthritis group. The ICI-induced inflammatory arthritis population and RA patients of European descent did not differ in frequency of having at least one SE allele, but ICI-induced inflammatory arthritis patients were more likely to be autoantibody-negative for RF and anti-CCP antibodies. CONCLUSION Patients with ICI-induced inflammatory arthritis of European descent were more likely to have at least one SE allele than healthy controls. Further studies are needed to validate these findings and investigate whether a unique immunogenetic framework increases risk for different immune-related adverse events.
Collapse
Affiliation(s)
- Laura C Cappelli
- Division of Rheumatology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mehmet T Dorak
- School of Life Sciences, Pharmacy & Chemistry, Kingston University London, London, UK
| | - Maria P Bettinotti
- Immunogenetics Laboratory, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Clifton O Bingham
- Division of Rheumatology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ami A Shah
- Division of Rheumatology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
20
|
Pistillo MP, Fontana V, Morabito A, Dozin B, Laurent S, Carosio R, Banelli B, Ferrero F, Spano L, Tanda E, Ferrucci PF, Martinoli C, Cocorocchio E, Guida M, Tommasi S, De Galitiis F, Pagani E, Antonini Cappellini GC, Marchetti P, Quaglino P, Fava P, Osella-Abate S, Ascierto PA, Capone M, Simeone E, Romani M, Spagnolo F, Queirolo P. Soluble CTLA-4 as a favorable predictive biomarker in metastatic melanoma patients treated with ipilimumab: an Italian melanoma intergroup study. Cancer Immunol Immunother 2019; 68:97-107. [PMID: 30311027 PMCID: PMC11028053 DOI: 10.1007/s00262-018-2258-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 10/04/2018] [Indexed: 12/11/2022]
Abstract
CTLA-4 blockade by means of ipilimumab (IPI) potentiates the immune response and improves overall survival (OS) in a minority of metastatic melanoma (MM) patients. We investigated the role of soluble CTLA-4 (sCTLA-4) as a possible biomarker for identifying this subset of patients. sCTLA-4 levels were analyzed at baseline in sera from 113 IPI-treated MM patients by ELISA, and the median value (200 pg/ml) was used to create two equally sized subgroups. Associations of sCTLA-4 with best overall response (BOR) to IPI and immune-related adverse events (irAEs) were evaluated through logistic regression. Kaplan-Meier and Cox regression methods were used to analyze OS. A remarkable association between sCTLA-4 levels and BOR was found. Specifically, the proportion of patients with sCTLA-4 > 200 pg/ml in irSD or irPD (immune-related stable or progressive disease) was, respectively, 80% (OR = 0.23; 95%CL = 0.03-1.88) and 89% (OR = 0.11; 95%CL = 0.02-0.71) and was lower than that observed among patients in irCR/irPR (immune-related complete/partial response). sCTLA-4 levels increased during IPI treatment, since the proportion of patients showing sCTLA > 200 pg/ml after 3 cycles was 4 times higher (OR = 4.41, 95%CL = 1.02-19.1) than that after 1 cycle. Moreover, a significantly lower death rate was estimated for patients with sCTLA-4 > 200 pg/ml (HR = 0.61, 95%CL = 0.39-0.98). Higher baseline sCTLA-4 levels were also associated with the onset of any irAE (p value = 0.029), in particular irAEs of the digestive tract (p value = 0.041). In conclusion, our results suggest that high sCTLA-4 serum levels might predict favorable clinical outcome and higher risk of irAEs in IPI-treated MM patients.
Collapse
Affiliation(s)
- Maria Pia Pistillo
- Unit of Tumor Epigenetics, IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi, 10, 16132, Genoa, Italy.
| | - Vincenzo Fontana
- Unit of Clinical Epidemiology, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Anna Morabito
- Unit of Tumor Epigenetics, IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi, 10, 16132, Genoa, Italy
| | - Beatrice Dozin
- Unit of Clinical Epidemiology, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Stefania Laurent
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Roberta Carosio
- Unit of Tumor Epigenetics, IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi, 10, 16132, Genoa, Italy
| | - Barbara Banelli
- Unit of Tumor Epigenetics, IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi, 10, 16132, Genoa, Italy
- Department of Health Sciences, University of Genoa, Genoa, Italy
| | - Francesca Ferrero
- Department of Medical Oncology, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Laura Spano
- Department of Medical Oncology, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Enrica Tanda
- Department of Medical Oncology, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | | | - Chiara Martinoli
- Oncology of Melanoma Unit, European Institute of Oncology, Milan, Italy
- iTeos Therapeutics, Gosselies, Belgium
| | | | - Michele Guida
- Department of Medical Oncology and Molecular Genetics Laboratory, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Stefania Tommasi
- Department of Medical Oncology and Molecular Genetics Laboratory, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | | | - Elena Pagani
- Istituto Dermopatico dell'Immacolata IDI-IRCCS, Rome, Italy
| | | | - Paolo Marchetti
- Istituto Dermopatico dell'Immacolata IDI-IRCCS, Rome, Italy
- Sapienza University of Rome, Rome, Italy
| | - Pietro Quaglino
- Department of Medical Sciences, Dermatologic Clinic, University of Turin, Turin, Italy
| | - Paolo Fava
- Department of Medical Sciences, Dermatologic Clinic, University of Turin, Turin, Italy
| | - Simona Osella-Abate
- Department of Medical Sciences, Section of Surgical Pathology, University of Turin, Turin, Italy
| | - Paolo Antonio Ascierto
- Melanoma, Cancer Immunotherapy and Innovative Therapy Unit, Istituto Nazionale Tumori Fondazione'G. Pascale', Naples, Italy
| | - Mariaelena Capone
- Melanoma, Cancer Immunotherapy and Innovative Therapy Unit, Istituto Nazionale Tumori Fondazione'G. Pascale', Naples, Italy
| | - Ester Simeone
- Melanoma, Cancer Immunotherapy and Innovative Therapy Unit, Istituto Nazionale Tumori Fondazione'G. Pascale', Naples, Italy
| | - Massimo Romani
- Unit of Tumor Epigenetics, IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi, 10, 16132, Genoa, Italy
| | - Francesco Spagnolo
- Department of Medical Oncology, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Paola Queirolo
- Department of Medical Oncology, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| |
Collapse
|
21
|
Romani M, Pistillo MP, Carosio R, Morabito A, Banelli B. Immune Checkpoints and Innovative Therapies in Glioblastoma. Front Oncol 2018; 8:464. [PMID: 30406030 PMCID: PMC6206227 DOI: 10.3389/fonc.2018.00464] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 10/02/2018] [Indexed: 12/26/2022] Open
Abstract
Targeting the Immune Checkpoint molecules (ICs; CTLA-4, PD-1, PD-L1/2, and others) which provide inhibitory signals to T cells, dramatically improves survival in hard-to-treat tumors. The establishment of an immunosuppressive environment prevents endogenous immune response in glioblastoma; therefore, manipulating the host immune system seems a reasonable strategy also for this tumor. In glioma patients the accumulation of CD4+/CD8+ T cells and Treg expressing high levels of CTLA-4 and PD-1, or the high expression of PD-L1 in glioma cells correlates with WHO high grade and short survival. Few clinical studies with IC inhibitors (ICis) were completed so far. Notably, the first large-scale randomized trial (NCT 02017717) that compared PD-1 blockade and anti-VEGF, did not show an OS increase in the patients treated with anti-PD-1. Several factors could have contributed to the failure of this trial and must be considered to design further clinical studies. In particular the possibility of targeting at the same time different ICs was pre-clinically tested in an animal model were inhibitors against IDO, CTLA-4 and PD-L1 were combined and showed persistent and significant antitumor effects in glioma-bearing mice. It is reasonable to hypothesize that the immunological characterization of the tumor in terms of type and level of expressed IC molecules on the tumor and TIL may be useful to design the optimal ICi combination for a given subset of tumor to overcome the immunosuppressive milieu of glioblastoma and to efficiently target a tumor with such high cellular complexity.
Collapse
Affiliation(s)
- Massimo Romani
- Laboratory of Tumor Epigenetics, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Maria Pia Pistillo
- Laboratory of Tumor Epigenetics, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Roberta Carosio
- Laboratory of Tumor Epigenetics, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Anna Morabito
- Laboratory of Tumor Epigenetics, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Barbara Banelli
- Laboratory of Tumor Epigenetics, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Health Sciences, University of Genova, Genova, Italy
| |
Collapse
|