1
|
Perrone G, Rigacci L, Roviello G, Landini I, Fabbri A, Iovino L, Puccini B, Cencini E, Orciuolo E, Bocchia M, Bosi A, Mini E, Nobili S. Validation of single nucleotide polymorphisms potentially related to R-CHOP resistance in diffuse large B-cell lymphoma patients. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:21. [PMID: 38835350 PMCID: PMC11149109 DOI: 10.20517/cdr.2024.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/30/2024] [Accepted: 05/16/2024] [Indexed: 06/06/2024]
Abstract
Aim: Diffuse large B-cell lymphoma (DLBCL) is the most common B-cell non-Hodgkin lymphoma (NHL). Despite the availability of clinical and molecular algorithms applied for the prediction of prognosis, in up to 30%-40% of patients, intrinsic or acquired drug resistance occurs. Constitutional genetics may help to predict R-CHOP resistance. This study aimed to validate previously identified single nucleotide polymorphisms (SNPs) in the literature as potential predictors of R-CHOP resistance in DLBCL patients, SNPs. Methods: Twenty SNPs, involved in R-CHOP pharmacokinetics/pharmacodynamics or other pathobiological processes, were investigated in 185 stage I-IV DLBCL patients included in a multi-institution pharmacogenetic study to validate their previously identified correlations with resistance to R-CHOP. Results: Correlations between rs2010963 (VEGFA gene) and sex (P = 0.046), and rs1625895 (TP53 gene) and stage (P = 0.003) were shown. After multivariate analyses, a concordant effect (i.e., increased risk of disease progression and death) was observed for rs1883112 (NCF4 gene) and rs1800871 (IL10 gene). When patients were grouped according to the revised International Prognostic Index (R-IPI), both these SNPs further discriminated progression-free survival (PFS) and overall survival (OS) of the R-IPI-1-2 subgroup. Overall, patients harboring the rare allele showed shorter PFS and OS compared with wild-type patients. Conclusions: Two out of the 20 study SNPs were validated. Thus, these results support the role of previously identified rs1883112 and rs1800871 in predicting DLBCL resistance to R-CHOP and highlight their ability to further discriminate the prognosis of R-IPI-1-2 patients. These data point to the need to also focus on host genetics for a more comprehensive assessment of DLBCL patient outcomes in future prospective trials.
Collapse
Affiliation(s)
- Gabriele Perrone
- Department of Health Sciences, University of Florence, Florence 50139, Italy
| | - Luigi Rigacci
- Research Unit of Hematology, Department of Medicine and Surgery, Campus Biomedico University, Rome 00128, Italy
| | | | - Ida Landini
- Department of Health Sciences, University of Florence, Florence 50139, Italy
| | - Alberto Fabbri
- Unit of Hematology, Azienda Ospedaliera Universitaria Senese, University of Siena, Siena 53100, Italy
| | - Lorenzo Iovino
- Unit of Hematology, Santa Chiara University Hospital, University of Pisa, Pisa 56126, Italy
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA 98109-4433, USA
| | - Benedetta Puccini
- Unit of Hematology, Careggi University-Hospital, Florence 50134, Italy
| | - Emanuele Cencini
- Unit of Hematology, Azienda Ospedaliera Universitaria Senese, University of Siena, Siena 53100, Italy
| | - Enrico Orciuolo
- Unit of Hematology, Santa Chiara University Hospital, University of Pisa, Pisa 56126, Italy
| | - Monica Bocchia
- Unit of Hematology, Azienda Ospedaliera Universitaria Senese, University of Siena, Siena 53100, Italy
| | - Alberto Bosi
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy
| | - Enrico Mini
- Department of Health Sciences, University of Florence, Florence 50139, Italy
- Authors contributed equally
| | - Stefania Nobili
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence 50139, Italy
- Authors contributed equally
| |
Collapse
|
2
|
Xin Z, You L, Li J, Na F, Chen M, Song J, Bai L, Chen H, Zhai J, Zhou X, Zhou J, Ying B. Immunogenetic polymorphisms predict therapeutic efficacy and survival outcomes in tumor patients receiving PD-1/PD-L1 blockade. Int Immunopharmacol 2023; 121:110469. [PMID: 37311357 DOI: 10.1016/j.intimp.2023.110469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/05/2023] [Accepted: 06/06/2023] [Indexed: 06/15/2023]
Abstract
BACKGROUND While immune checkpoint inhibitors (ICIs) demonstrate remarkable clinical responses, only a small subset of patients obtains benefits. Genes linked to the tumor immune system are confirmed to be critical for the treatment of ICIs, and their polymorphisms can contribute to ICI efficacy. Here, we examined the potential of immunogenetic variations to predict the efficacy and survival of the PD-1/PD-L1 blockade. METHODS Cancerous patients receiving PD-1/PD-L1 blockade were recruited and followed up. Pivotal genes related to tumor-immunity were filtered through a protein-protein interaction network and the degree algorithm in Cytoscape. Finally, 39 genetic variants were genotyped through multiplex genotyping assays. Association analyses between variants and ICI efficacy and progression-free survival (PFS) were performed. RESULTS Overall, 318 patients were ultimately enrolled. Hence, three immunogenetic variants were identified as predictors of PD-1/PD-L1 blockade response. Mutant alleles from ATG7 rs7625881, CD274 rs2297136, and TLR4 rs1927911 were all at increased risk of tumor progression following ICI therapy (OR: 1.475, 1.641, 1.462, respectively; P value: 0.028, 0.017, 0.027, respectively). Significant immunogenetic variants also attained similar trends in the PD-1 blockade, lung cancer, or lung cancer using PD-1 blockade subgroups. Furthermore, the mutant genotypes of CD274 rs2297136 (GG as the reference: HR: 0.50 (95%CI: 0.29-0.88), P value: 0.015) and TLR4 rs1927911 (AA as the reference: HR: 0.65 (95%CI: 0.47-0.91), P value: 0.012) indicated poorer PFS and were both independent prognostic factors. CONCLUSION Immunogenetic polymorphisms, including ATG7 rs7625881, CD274 rs2297136, and TLR4 rs1927911, were first identified as potential predictors of response to PD-1/PD-L1 blockade in tumor patients.
Collapse
Affiliation(s)
- Zhaodan Xin
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, PR China
| | - Liting You
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, PR China; Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, PR China
| | - Jin Li
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, PR China
| | - Feifei Na
- Department of Thoracic Cancer, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, PR China
| | - Min Chen
- Department of Laboratory Medicine, The First Affiliated Hospital of Hainan Medical College, Haikou, Hainan Province 570102, PR China
| | - Jiajia Song
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, PR China
| | - Ling Bai
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, PR China
| | - Hao Chen
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, PR China
| | - Jianzhao Zhai
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, PR China
| | - Xiaohan Zhou
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, PR China
| | - Juan Zhou
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, PR China.
| | - Binwu Ying
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, PR China.
| |
Collapse
|
3
|
Shihong Gao D, Zhu X, Lu B. Development and application of sensitive, specific, and rapid CRISPR-Cas13-based diagnosis. J Med Virol 2021; 93:4198-4204. [PMID: 33599292 PMCID: PMC8014745 DOI: 10.1002/jmv.26889] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 01/15/2021] [Accepted: 01/20/2021] [Indexed: 12/22/2022]
Abstract
Nucleic acid detection is a necessary part of medical treatment and fieldwork. However, the current detection technologies are far from ideal. A lack of timely and accessible testing for identifying cases and close contacts has allowed severe acute respiratory syndrome coronavirus‐2 (SARS‐CoV‐2), the causative virus of the ongoing coronavirus disease‐2019 (COVID‐19) pandemic, to spread uncontrollably. The slow and expensive detection of mutations—predictors for chronic diseases such as cancer—form a barrier to personalized treatment. A recently developed diagnostic assay is ideal and field‐ready—it relies on CRISPR‐Cas13. CRISPR‐Cas13 works similarly to other CRISPR systems: Cas13 is guided by a crRNA to cleave next to a specific RNA target sequence. Additionally, Cas13 boasts a unique collateral cleavage activity; collateral cleavage of a fluorescent reporter detects the presence of the target sequence in sample RNA. This system forms the basis of CRISPR‐Cas13 diagnostic assays. CRISPR‐Cas13 assays have >95% sensitivity and >99% specificity. Detection is rapid (<2 h), inexpensive ($0.05 per test), and portable—a test using lateral flow strips is akin to a pregnancy test. The recent adaptation of micro‐well chips facilitates high‐level multiplexing and is high‐throughput. In this review, we cover the development of CRISPR‐Cas13 assays for medical diagnosis, discuss the advantages of CRISPR‐Cas13‐based diagnosis over the traditional reverse transcription polymerase chain reaction (RT‐PCR), and present examples of detection from real patient samples. In the CRISPR‐Cas13 system, Cas13 is guided by a crRNA to cleave sample RNA at a specific target sequence. Cas13 has collateral cleavage activity that when used with a fluorescent reporter detects a specific target sequence. CRISPR‐Cas13‐based diagnostics are highly sensitive (>95%), specific (>99%), rapid (<2 h), inexpensive ($0.05 per test), and portable‐recent adaptions of micro‐well chips facilitate high‐evel multiplexing and are high‐throughput. CRISPR‐Cas13‐based diagnostics have been validated in the detection of viral RNA and mutations in patient samples, including SARS‐CoV‐2 RNA during the ongoing COVID‐19 pandemic. CRISPR‐Cas13 is also applied to RNA editing, thereby creating a full system of diagnostics and therapeutics.
Collapse
Affiliation(s)
- David Shihong Gao
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Xiaodong Zhu
- Department of Biological Sciences, Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Binfeng Lu
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
4
|
Milano G, Innocenti F, Ciccolini J. The association between adverse events and outcome under checkpoint inhibitors: Where is the deal? Transl Oncol 2020; 14:100952. [PMID: 33260071 PMCID: PMC7708939 DOI: 10.1016/j.tranon.2020.100952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/30/2020] [Accepted: 11/09/2020] [Indexed: 11/22/2022] Open
Abstract
A review which lays out different potential contributions which can help to understand the IRAEs-outcome link. There is a possibility to compute a multifactorial index to characterise patients as ICI sensitive or ICI unsensitive. Prospective trails with ICIs are now fesaible to shape patient care beyond high -dose steroids.
Recent reports have put into evidence the possibility of a link between immune-related adverse events (IRAEs) and treatment outcome, patients drawing a benefit from treatment being also exposed to the risk to develop toxicity. A still unanswered question remains the biological origin(s) which can sustain and explain such a relationship. The purpose of this review paper is to lay out different potential contributions which can help to understand the IRAEs-outcome link and to propose clinical perspectives taking advantage of this association. In this respect, pharmacokinetics aspects, immunological and immunogenetics implications have been taken into consideration.
Collapse
Affiliation(s)
- Gerard Milano
- UNS EA 7497 Nice University, Centre Antoine Lacassagne, 33 avenue de Valombrose, 06189 Cedex 2, France.
| | - Federico Innocenti
- UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
| | | |
Collapse
|
5
|
Biological Factors behind Melanoma Response to Immune Checkpoint Inhibitors. Int J Mol Sci 2020; 21:ijms21114071. [PMID: 32517213 PMCID: PMC7313051 DOI: 10.3390/ijms21114071] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/01/2020] [Accepted: 06/02/2020] [Indexed: 12/14/2022] Open
Abstract
Modern immunotherapy together with targeted therapy has revolutionized the treatment of advanced melanoma. Inhibition of immune checkpoints significantly improved the median overall survival and gave hope to many melanoma patients. However, this treatment has three serious drawbacks: high cost, serious side effects, and an effectiveness limited only to approximately 50% of patients. Some patients do not derive any or short-term benefit from this treatment due to primary or secondary resistance. The response to immunotherapy depends on many factors that fall into three main categories: those associated with melanoma cells, those linked to a tumor and its microenvironment, and those classified as individual ontogenic and physiological features of the patient. The first category comprises expression of PD-L1 and HLA proteins on melanoma cells as well as genetic/genomic metrics such as mutational load, (de)activation of specific signaling pathways and epigenetic factors. The second category is the inflammatory status of the tumor: “hot” versus “cold” (i.e., high versus low infiltration of immune cells). The third category comprises metabolome and single nucleotide polymorphisms of specific genes. Here we present up-to-date data on those biological factors influencing melanoma response to immunotherapy with a special focus on signaling pathways regulating the complex process of anti-tumor immune response. We also discuss their potential predictive capacity.
Collapse
|
6
|
Abstract
Cancer immunotherapy is based on checkpoint inhibitors (CPIs) that significantly improve the clinical outcome of several malignant diseases. These inhibitors are monoclonal antibodies (mAbs) directed at cytotoxic T lymphocyte-associated protein 4 (CTLA-4), programmed cell death 1 (PD-1), or programmed death-ligand 1 (PD-L1), sharing most of the clinical pharmacokinetic characteristics of mAb targeted therapies, all of which differ from those of cytotoxics and small molecules. Establishing the labeled dose of mAbs, and particularly of the CPIs, represents a true challenge. This review therefore examines the main criteria used for dose selection, along with their limits. The relationships between CPI pharmacokinetic parameters and treatment outcome (efficacy and/or toxicity) differ somewhat among the various drugs, but general features can be identified. Nevertheless, the interpretation of these relationships remains quite controversial. A first interpretation asserts that inter-individual pharmacokinetic variability in clearance has an impact on outcome and should be taken into consideration for dosing individualization. The second considers that higher clearance values observed in some patients result from characteristics associated with poor predictive factors of efficacy. Finally, the schedule, and particularly its frequency of administration, merits rethinking.
Collapse
|