1
|
Chen L, Cai B, Ni X, Lin Q, Ke R, Wan X, Huang T, Shan X, Wang B. Temozolomide monotherapy versus combination therapies in melanoma: a meta-analysis of efficacy and safety. Melanoma Res 2025; 35:87-101. [PMID: 39874124 DOI: 10.1097/cmr.0000000000001021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2025]
Abstract
Temozolomide is used in melanoma therapy, but the comparative efficacy and safety of monotherapy vs combination therapies are unclear. This meta-analysis evaluates temozolomide monotherapy vs combination therapies in melanoma patients. PubMed, Embase, and Cochrane Library were searched up to August 2024 for studies comparing temozolomide monotherapy with combination therapies in melanoma. Primary outcomes were 1-year survival and objective response rates (RR); secondary outcomes included hematologic and non-hematologic toxicities. Data were pooled using risk ratios with 95% confidence intervals (CIs). Seven studies were included. Combination therapies improved objective RR over temozolomide monotherapy (risk ratio 0.68, 95% CI: 0.53-0.88). One-year survival did not differ significantly between groups (risk ratio 0.81, 95% CI: 0.59-1.12). Temozolomide monotherapy was associated with reduced incidence of leukopenia (risk ratio 0.54, 95% CI: 0.30-0.95). Adding interferon-alpha (IFN-α) to temozolomide significantly improved 1-year survival (risk ratio 0.54, 95% CI: 0.35-0.84) and objective RR (risk ratio 0.57, 95% CI: 0.42-0.78) compared to temozolomide alone, without significantly increasing toxicity. Combination therapies enhance objective RR over temozolomide monotherapy, with similar 1-year survival. Temozolomide monotherapy offers a better hematologic safety profile. Combining temozolomide with IFN-α significantly improves survival and RR without increasing toxicity. Clinicians should balance efficacy and safety when choosing melanoma treatments.
Collapse
Affiliation(s)
- Lu Chen
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Beichen Cai
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Xuejun Ni
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Qian Lin
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Ruonan Ke
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Xiaofen Wan
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Tao Huang
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Xiuying Shan
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Biao Wang
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| |
Collapse
|
2
|
Khan SR, Breadner D. Unveiling the Synergistic Potential: Bispecific Antibodies in Conjunction with Chemotherapy for Advanced Non-Small-Cell Lung Cancer Treatment. Curr Oncol 2025; 32:206. [PMID: 40277763 PMCID: PMC12025875 DOI: 10.3390/curroncol32040206] [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: 02/28/2025] [Revised: 03/23/2025] [Accepted: 03/27/2025] [Indexed: 04/26/2025] Open
Abstract
Lung cancer remains the leading cause of cancer-related mortality worldwide, with non-small-cell lung cancer (NSCLC) accounting for the majority of the cases. Despite advancements in targeted therapies and immunotherapies, many patients still rely on chemotherapy, highlighting the need for innovative treatment strategies. Bispecific antibodies (bsAbs), which feature two distinct binding sites capable of targeting different antigens, have emerged as a promising therapeutic approach, particularly in combination with chemotherapy. This review explores the scientific evolution and clinical application of bsAbs in NSCLC, focusing on their synergistic potential with chemotherapy. BsAbs, such as amivantamab, which targets EGFR and MET, have demonstrated significant efficacy in clinical trials, particularly in patients with EGFR mutations. The combination of bsAbs with chemotherapy enhances immune-mediated tumor destruction by modulating the tumor microenvironment and overcoming resistance mechanisms. Recent clinical trials have shown improved progression-free survival and overall survival when bsAbs such as amivantamab are combined with chemotherapy, underscoring their potential to transform NSCLC treatment. Many other clinical trials are underway that are evaluating newer bsAbs, such as ivonescimab, which targets PD1 and VEGF. This review also discusses ongoing clinical trials investigating various bsAbs targeting EGFR, PD-1, PD-L1, HER2, and other pathways, highlighting the future directions of bsAb-based therapies. As the field evolves, bsAbs are poised to become a cornerstone of multimodal NSCLC treatment, offering more effective and personalized therapeutic options for patients with advanced disease.
Collapse
Affiliation(s)
- Saqib Raza Khan
- Division of Medical Oncology, Department of Oncology, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 3K7, Canada;
- Verspeeten Family Cancer Centre, London Health Sciences Centre, London, ON N6A 5W9, Canada
| | - Daniel Breadner
- Division of Medical Oncology, Department of Oncology, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 3K7, Canada;
- Verspeeten Family Cancer Centre, London Health Sciences Centre, London, ON N6A 5W9, Canada
| |
Collapse
|
3
|
Zannoni GF, Angelico G, Spadola S, Bragantini E, Troncone G, Fraggetta F, Santoro A. Chemotherapy Response Score (CRS): A comprehensive review of its prognostic and predictive value in High-Grade Serous Carcinoma (HGSC). Gynecol Oncol 2025; 194:1-10. [PMID: 39919553 DOI: 10.1016/j.ygyno.2025.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 01/25/2025] [Accepted: 01/28/2025] [Indexed: 02/09/2025]
Abstract
Ovarian carcinoma, the second most common gynecological cancer in Western countries, is frequently diagnosed at advanced stages, necessitating complex treatment strategies. While cytoreductive surgery remains the standard for improving survival, neoadjuvant chemotherapy (NACT) has become essential for cases unsuitable for immediate surgery, aiming to reduce tumor burden preoperatively. Introduced in 2015, the Chemotherapy Response Score (CRS) is now a key histopathological tool for assessing response to NACT, stratifying patients into three response categories. CRS3 is associated with improved progression-free survival (PFS) and overall survival (OS), while CRS1 and CRS2 are linked to poorer outcomes. Validated across clinical cohorts, CRS has proven valuable not only as a prognostic tool but also as a predictor for molecular-targeted therapies, such as PARP inhibitors, especially in BRCA wild-type patients. Studies also suggest a potential role for CRS in guiding the use of PD-L1 inhibitors, especially in partial responders (CRS1 and CRS2), where immunotherapy may complement chemotherapy. In the present paper we exlored the actual knowledge on CRS scoring for ovarian carcinoma. Diagnostic and prognostic implications of CRS as well as its correlation with therapeutic response and other biomarkers are discussed.
Collapse
Affiliation(s)
- Gian Franco Zannoni
- Pathology Unit, Department of Woman and Child's Health and Public Health Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; Pathology Institute, Catholic University of Sacred Heart, 00168 Rome, Italy.
| | - Giuseppe Angelico
- Department of Medicine and Surgery, Kore University of Enna, 94100 Enna, Italy
| | - Saveria Spadola
- Department of Medicine and Surgery, Kore University of Enna, 94100 Enna, Italy
| | - Emma Bragantini
- Department of Surgical Pathology, Ospedale S. Chiara 9, 38122 Trento, Italy
| | - Giancarlo Troncone
- Department of Public Health, University of Naples, "Federico II", Naples, Italy
| | | | - Angela Santoro
- Pathology Unit, Department of Woman and Child's Health and Public Health Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| |
Collapse
|
4
|
Martin-Aubert S, Avrillon K, Tournier N, Bordat A, Tran VL, Ibrahim N, Kereselidze D, Jego B, Potiron L, Tsapis N, Nicolas J, Boissenot T, Truillet C. Successful repositioning of mertansine for improved chemotherapy by combining a polymer prodrug approach and PET imaging. J Control Release 2025; 378:803-813. [PMID: 39719212 DOI: 10.1016/j.jconrel.2024.12.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 12/01/2024] [Accepted: 12/19/2024] [Indexed: 12/26/2024]
Abstract
Mertansine (DM1), a potent tumor-killing maytansinoid, requires conjugation to antibodies or incorporation into nanocarriers due to its high toxicity. However, these carriers often result in undesirable biodistribution, leading to rapid and long-term accumulation in the kidneys or liver and potentially increased toxicity. To overcome this limitation, we used the hydrophilic, biocompatible, and stealth properties of polyacrylamide (PAAm) as a scaffold to develop water-soluble PAAm-DM1 polymer prodrugs, leveraging PAAm's previous success in delivering paclitaxel via subcutaneous administration. To monitor distribution and predict efficacy, we have imparted Positron Emission Tomography (PET) imaging capabilities to well-defined PAAm-DM1 polymer prodrugs. Our studies demonstrated the same tumor accumulation and the same distribution of PAAm-DM1 in the main organs such as liver, kidneys muscle, regardless of delivery route (subcutaneous or intravenous). Interestingly, tumor accumulation of PAAm-DM1 was primarily driven by passive accumulation, as indicated by PET imaging, without significantly altering treatment efficacy. This suggests complex mechanisms, possibly involving immune system interactions by influencing notably the metabolism and clearance. To enhance therapeutic outcomes, we combined the polymer prodrug with immunotherapy, specifically anti-CTLA4. Our findings highlight the promising potential of PAAm-DM1, offering a novel formulation strategy for DM1 in cancer therapy.
Collapse
Affiliation(s)
- Soizic Martin-Aubert
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, SHFJ, Orsay 91401, France..
| | - Kevin Avrillon
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, SHFJ, Orsay 91401, France
| | - Nicolas Tournier
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, SHFJ, Orsay 91401, France..
| | | | - Vu Long Tran
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, SHFJ, Orsay 91401, France
| | - Nada Ibrahim
- Imescia, Université Paris-Saclay, 91400 Saclay, France
| | - Dimitri Kereselidze
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, SHFJ, Orsay 91401, France
| | - Benoit Jego
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, SHFJ, Orsay 91401, France..
| | - Léa Potiron
- Imescia, Université Paris-Saclay, 91400 Saclay, France
| | - Nicolas Tsapis
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400 Orsay, France.
| | - Julien Nicolas
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400 Orsay, France.
| | | | - Charles Truillet
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, SHFJ, Orsay 91401, France..
| |
Collapse
|
5
|
Jiang C, Hong Z, Liu S, Hong Z, Dai B. Roles of CDK12 mutations in PCa development and treatment. Biochim Biophys Acta Rev Cancer 2025; 1880:189247. [PMID: 39681197 DOI: 10.1016/j.bbcan.2024.189247] [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: 07/14/2024] [Revised: 12/05/2024] [Accepted: 12/11/2024] [Indexed: 12/18/2024]
Abstract
Prostate cancer (PCa) is one of the most common cancers in men, and cyclin-dependent kinase 12 (CDK12) is emerging as a novel star player in the PCa tumorigenesis and progression to castration-resistant prostate cancer (CRPC). In PCa, CDK12 alterations are mostly loss-of-function mutations featuring intronic polyadenylation (IPA), focal tandem duplications (FTDs), and R-loops formation and transcription-replication conflicts (TRCs). The occurrence of IPA can result in homologous recombination deficiency (HRD) and androgen receptor (AR) variation. FTDs induce neoantigens and increase the expression of the AR, MYC, and other hotspot- associated genes. R-loops lead to TRCs and influence various cellular processes, including gene expression and genome stability. Due to the poor prognosis of CDK12-mutant PCa patients and the mediocre response to classic standard therapies, HRD and increased neoantigen levels have provided clinicians with new insights into alternative systematic treatments for this novel PCa phenotype. In this review, we summarize the roles of CDK12 mutations in PCa and discuss their clinical value, suggesting that CDK12 potentially represents a target for further research and the development of clinical strategies for PCa.
Collapse
Affiliation(s)
- Chenye Jiang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Genitourinary Cancer Institute, Shanghai 200032, China
| | - Zhe Hong
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Genitourinary Cancer Institute, Shanghai 200032, China.
| | - Shiwei Liu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Genitourinary Cancer Institute, Shanghai 200032, China
| | - Zongyuan Hong
- Laboratory of Quantitative Pharmacology, Wannan Medical College, Wuhu 241002, China
| | - Bo Dai
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Genitourinary Cancer Institute, Shanghai 200032, China.
| |
Collapse
|
6
|
de Moraes FCA, Sano VKT, Silva BL, Silva ALS, Castro SCR, Kreuz M, Fernandes LR, Kelly FA, Burbano RMR. PD-1/PD-L1 Inhibitors Increase Pathological Complete Response in Locally Advanced Gastric Cancer: A Meta-analysis and Trial Sequential Analysis. J Gastrointest Cancer 2025; 56:49. [PMID: 39833372 DOI: 10.1007/s12029-024-01141-4] [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] [Accepted: 11/07/2024] [Indexed: 01/22/2025]
Abstract
BACKGROUND AND OBJECTIVE Gastric cancer (GC) remains a leading cause of morbidity and mortality worldwide. The current standard of care involves neoadjuvant chemotherapy (NACT) followed by radical gastrectomy. This study aims to evaluate the efficacy of neoadjuvant therapy with PD-1/PD-L1 inhibitors in comparison to chemotherapy alone for patients with locally advanced gastric cancer (LAGC). METHODS We conducted a systematic search of PubMed, Web of Science, and Embase to identify studies examining the addition of PD-1/PD-L1 inhibitors to neoadjuvant therapy for LAGC. Odds ratios (OR) were calculated for binary outcomes, such as pathological complete response (pCR), with corresponding 95% confidence intervals (CI). RESULTS Seven studies were included, encompassing a total of 1772 patients. Baseline median age ranged from 31 to 75 years. Most patients had an ECOG performance status score of 0 (942 patients), while 294 had an ECOG score of 1. The estimated pCR (OR 5.94, 95% CI 3.98-8.87; p < 0.000001) significantly favored the PD-1/PD-L1 inhibitors combined with chemotherapy over chemotherapy alone. Additionally, the incidence of certain adverse events increased significantly in the intervention group, including any-grade hypothyroidism (OR 4.55, 95% CI 2.27-9.10; p = 0.000019) and rash (OR 1.74, 95% CI 1.10-2.76; p = 0.017). Conversely, the control group showed a statistically significant lower incidence of grade ≥ 3 fatigue (OR 2.80, 95% CI 1.15-6.85; p = 0.024) compared to the intervention group. CONCLUSION This systematic review and meta-analysis indicate that the addition of PD-1/PD-L1 inhibitors to neoadjuvant chemotherapy is associated with a higher pathological complete response rate compared to chemotherapy alone in patients with locally advanced gastric cancer.
Collapse
Affiliation(s)
| | | | - Barbara Lins Silva
- Vancouver Island Health Authority, 1947 Cook St, Victoria, BC, V8T 3P7, Canada
| | | | | | - Michele Kreuz
- Lutheran University of Brazil, Rio Grande Do Sul, Canoas, 92425-020, Brazil
| | | | | | | |
Collapse
|
7
|
Woo JW, Han EK, Suh KJ, Kim SH, Kim JH, Park SY. Alteration of PD-L1 (SP142) status after neoadjuvant chemotherapy and its clinical significance in triple-negative breast cancer. Breast Cancer Res Treat 2024; 207:301-311. [PMID: 38753066 PMCID: PMC11297096 DOI: 10.1007/s10549-024-07359-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 04/24/2024] [Indexed: 08/03/2024]
Abstract
PURPOSE The tumor immune microenvironment can change after neoadjuvant chemotherapy (NAC) for triple-negative breast cancer (TNBC). We aimed to investigate the effects of NAC on PD-L1 (SP142) status and its clinical significance in TNBC. METHODS Paired samples of biopsy and resection specimens were collected from 182 patients with TNBC before and after NAC. PD-L1 (SP142) expression in immune cells in pre- and post-NAC breast cancer samples and the changes between them were analyzed, along with their relationships with the clinicopathological features and clinical outcomes of the patients. RESULTS Of the 182 patients, 61 (33.5%) achieved pathologic complete response (pCR) after NAC. PD-L1 (SP142) positivity, defined as immune cell staining in ≥ 1% of tumor area, was a predictor for pCR. PD-L1-positive immune cells significantly increased after NAC (2.8% to 5.2% on average) in 109 patients with measurable residual disease. Alteration of PD-L1 status was observed in 24 (22.0%) of the 109 patients with measurable residual tumors after NAC, and all PD-L1 status-converted patients, except one, revealed negative-to-positive conversion. Regarding chemotherapeutic agents, the use of platinum agents was associated with a significant increase in PD-L1-positive immune cells after NAC. In survival analyses, a positive PD-L1 status after NAC and increase of PD-L1-positive immune cells after NAC were associated with better recurrence-free survival of the patients. CONCLUSION PD-L1 (SP142) status changes after NAC, mostly as a positive conversion. As PD-L1 (SP142) status can convey prognostic and predictive information, it needs to be tested before and after NAC.
Collapse
Affiliation(s)
- Ji Won Woo
- Department of Pathology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, 82, Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam, Gyeonggi, 13620, Republic of Korea
| | - Eun Kyung Han
- Department of Pathology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, 82, Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam, Gyeonggi, 13620, Republic of Korea
| | - Koung Jin Suh
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Gyeonggi, Republic of Korea
| | - Se Hyun Kim
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Gyeonggi, Republic of Korea
| | - Jee Hyun Kim
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Gyeonggi, Republic of Korea
| | - So Yeon Park
- Department of Pathology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, 82, Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam, Gyeonggi, 13620, Republic of Korea.
| |
Collapse
|
8
|
Chen L, Zhu W, Zhang W, Chen E, Zhou W. Magnetic resonance imaging radiomics-based prediction of severe inflammatory response in locally advanced rectal cancer patients after neoadjuvant radiochemotherapy. Langenbecks Arch Surg 2024; 409:218. [PMID: 39017754 PMCID: PMC11255083 DOI: 10.1007/s00423-024-03416-7] [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: 10/19/2023] [Accepted: 07/12/2024] [Indexed: 07/18/2024]
Abstract
PURPOSE To predict severe inflammatory response after neoadjuvant radiochemotherapy in locally advanced rectal cancer (RC) patients using magnetic resonance imaging (MRI) radiomics models. METHODS This retrospective study included patients who underwent radical surgery for RC cancer after neoadjuvant radiochemotherapy between July 2017 and December 2019 at XXX Hospital. MRI radiomics features were extracted from T2WI images before (pre-nRCT-RF) and after (post-nRCT-RF) neoadjuvant radiochemotherapy, and the variation of radiomics features before and after neoadjuvant radiochemotherapy (delta-RF) were calculated. Eight, eight, and five most relevant features were identified for pre-nRCT-RF, post-nRCT-RF, and delta-RF, respectively. RESULTS Eighty-six patients were included and randomized 3:1 to the training and test set (n = 65 and n = 21, respectively). The prediction model based on delta-RF had areas under the curve (AUCs) of 0.80 and 0.85 in the training and test set, respectively. A higher rate of difficult operations was observed in patients with severe inflammation (65.5% vs. 42.9%, P = 0.045). CONCLUSION The prediction model based on MRI delta-RF may be a useful tool for predicting severe inflammatory response after neoadjuvant radiochemotherapy in locally advanced RC patients.
Collapse
Affiliation(s)
- Li Chen
- Department of Colorectal Surgery, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China.
| | - Wenchao Zhu
- Department of Radiology, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Wei Zhang
- Department of Colorectal Surgery, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Engeng Chen
- Department of Colorectal Surgery, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Wei Zhou
- Department of Colorectal Surgery, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| |
Collapse
|
9
|
Cheng KW, Yen CH, Chang R, Wei JCC, Wang SI. Real-World Assessment of Recommended COVID-19 Vaccination Waiting Period after Chemotherapy. Vaccines (Basel) 2024; 12:678. [PMID: 38932407 PMCID: PMC11209144 DOI: 10.3390/vaccines12060678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 06/14/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024] Open
Abstract
There is a knowledge gap concerning the proper timing for COVID-19 vaccination in cancer patients undergoing chemotherapy. We aimed to evaluate the suitability of the guidelines that recommend waiting at least three months after undergoing chemotherapy before receiving a COVID-19 vaccine. This retrospective cohort study used aggregated data from the TriNetX US Collaboratory network. Participants were grouped into two groups based on the interval between chemotherapy and vaccination. The primary outcome assessed was infection risks, including COVID-19; skin, intra-abdominal, and urinary tract infections; pneumonia; and sepsis. Secondary measures included healthcare utilization and all causes of mortality. Kaplan-Meier analysis and the Cox proportional hazard model were used to calculate the cumulative incidence and hazard ratio (HR) and 95% confidence intervals for the outcomes. The proportional hazard assumption was tested with the generalized Schoenfeld approach. Four subgroup analyses (cancer type, vaccine brand, sex, age) were conducted. Sensitivity analyses were performed to account for competing risks and explore three distinct time intervals. Patients receiving a vaccine within three months after chemotherapy had a higher risk of COVID-19 infection (HR: 1.428, 95% CI: 1.035-1.970), urinary tract infection (HR: 1.477, 95% CI: 1.083-2.014), and sepsis (HR: 1.854, 95% CI: 1.091-3.152) compared to those who adhered to the recommendations. Hospital inpatient service utilization risk was also significantly elevated for the within three months group (HR: 1.692, 95% CI: 1.354-2.115). Adhering to a three-month post-chemotherapy waiting period reduces infection and healthcare utilization risks for cancer patients receiving a COVID-19 vaccine.
Collapse
Affiliation(s)
- Kai-Wen Cheng
- Department of Emergency Medicine, China Medical University Hospital, Taichung 40447, Taiwan;
| | - Chi-Hua Yen
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan;
- Department of Family and Community Medicine, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| | - Renin Chang
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 81362, Taiwan;
- Department of Recreation and Sports Management, Tajen University, Pintung 90741, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
| | - James Cheng-Chung Wei
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- Department of Nursing, Chung Shan Medical University, Taichung 40201, Taiwan
- Department of Allergy, Immunology & Rheumatology, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
- Graduate Institute of Integrated Medicine, China Medical University, Taichung 40447, Taiwan
- Office of Research and Development, Asia University, Taichung 41354, Taiwan
| | - Shiow-Ing Wang
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- Center for Health Data Science, Department of Medical Research, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| |
Collapse
|
10
|
Yee EJ, Gilbert D, Kaplan J, Wani S, Kim SS, McCarter MD, Stewart CL. Effect of Neoadjuvant Chemotherapy on Tumor-Infiltrating Lymphocytes in Resectable Gastric Cancer: Analysis from a Western Academic Center. Cancers (Basel) 2024; 16:1428. [PMID: 38611107 PMCID: PMC11010931 DOI: 10.3390/cancers16071428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 03/29/2024] [Accepted: 04/06/2024] [Indexed: 04/14/2024] Open
Abstract
Tumor-infiltrating lymphocytes (TILs) are an emerging biomarker predictive of response to immunotherapy across a spectrum of solid organ malignancies. The characterization of TILs in gastric cancer (GC) treated with contemporary, multiagent neoadjuvant chemotherapy (NAC) is understudied. In this retrospective investigation, we analyzed the degree of infiltration, phenotype, and spatial distribution of TILs via immunohistochemistry within resected GC specimens treated with or without NAC at a Western center. We hypothesized that NAC executes immunostimulatory roles, as evidenced by an increased number of anti-tumor TILs in the tumor microenvironment. We found significantly elevated levels of conventional and memory CD8+ T cells, as well as total TILs (CD4+, CD8+, Treg, B cells), within chemotherapy-treated tumors compared with chemotherapy-naïve specimens. We also revealed important associations between survival and pathologic responses with enhanced TIL infiltration. Taken together, our findings advocate for an immunostimulatory role of chemotherapy and underscore the potential synergistic effect of combining chemotherapy with immunotherapy in resectable gastric cancer.
Collapse
Affiliation(s)
- Elliott J. Yee
- Division of Surgical Oncology, Department of Surgery, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; (D.G.); (M.D.M.); (C.L.S.)
| | - Danielle Gilbert
- Division of Surgical Oncology, Department of Surgery, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; (D.G.); (M.D.M.); (C.L.S.)
| | - Jeffrey Kaplan
- Department of Pathology, University of Colorado, Aurora, CO 80045, USA;
| | - Sachin Wani
- Division of Gastroenterology, Department of Medicine, University of Colorado, Aurora, CO 80045, USA;
| | - Sunnie S. Kim
- Division of Medical Oncology, Department of Medicine, University of Colorado, Aurora, CO 80045, USA;
| | - Martin D. McCarter
- Division of Surgical Oncology, Department of Surgery, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; (D.G.); (M.D.M.); (C.L.S.)
| | - Camille L. Stewart
- Division of Surgical Oncology, Department of Surgery, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; (D.G.); (M.D.M.); (C.L.S.)
| |
Collapse
|
11
|
Wickenberg M, Mercier R, Yap M, Walker J, Baker K, LaPointe P. Hsp90 inhibition leads to an increase in surface expression of multiple immunological receptors in cancer cells. Front Mol Biosci 2024; 11:1334876. [PMID: 38645275 PMCID: PMC11027010 DOI: 10.3389/fmolb.2024.1334876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 03/20/2024] [Indexed: 04/23/2024] Open
Abstract
Heat shock protein 90 (Hsp90) is a molecular chaperone important for maintaining protein homeostasis (proteostasis) in the cell. Hsp90 inhibitors are being explored as cancer therapeutics because of their ability to disrupt proteostasis. Inhibiting Hsp90 increases surface density of the immunological receptor Major Histocompatibility Complex 1 (MHC1). Here we show that this increase occurs across multiple cancer cell lines and with both cytosol-specific and pan-Hsp90 inhibitors. We demonstrate that Hsp90 inhibition also alters surface expression of both IFNGR and PD-L1, two additional immunological receptors that play a significant role in anti-tumour or anti-immune activity in the tumour microenvironment. Hsp90 also negatively regulates IFN-γ activity in cancer cells, suggesting it has a unique role in mediating the immune system's response to cancer. Our data suggests a strong link between Hsp90 activity and the pathways that govern anti-tumour immunity. This highlights the potential for the use of an Hsp90 inhibitor in combination with another currently available cancer treatment, immune checkpoint blockade therapy, which works to prevent immune evasion of cancer cells. Combination checkpoint inhibitor therapy and the use of an Hsp90 inhibitor may potentiate the therapeutic benefits of both treatments and improve prognosis for cancer patients.
Collapse
Affiliation(s)
- Madison Wickenberg
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Rebecca Mercier
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Megan Yap
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - John Walker
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Kristi Baker
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Paul LaPointe
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
12
|
Zhang M, Zhang G, Niu Y, Zhang G, Ji Y, Yan X, Zhang X, Wang Q, Jing X, Wang J, Ma Z, Wang H. Sintilimab with two cycles of chemotherapy for the treatment of advanced squamous non-small cell lung cancer: a phase 2 clinical trial. Nat Commun 2024; 15:1512. [PMID: 38374204 PMCID: PMC10876536 DOI: 10.1038/s41467-024-45769-z] [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: 08/10/2023] [Accepted: 02/05/2024] [Indexed: 02/21/2024] Open
Abstract
This was a single-arm, multicenter phase 2 clinical trial (ChiCTR1900021726) involving advanced squamous non-small cell lung cancer (sq-NSCLC) patients undergoing 2 cycles of nab-paclitaxel/carboplatin and sintilimab (anti-PD-1), followed by sintilimab maintenance therapy. The median progression-free survival (PFS) was 11.4 months (95% CI: 6.7-18.1), which met the pre-specified primary endpoint. Secondary endpoints included objective response rate reaching 70.5% and a disease control rate of 93.2%, with a median duration of response of 13.6 months [95% CI: 7.0-not evaluable (NE)]. The median overall survival was 27.2 months (95% CI: 20.2-NE) with treatment-related adverse events grades ≥3 occurring in 10.9% of patients. Predefined exploratory endpoints comprised relationships between biomarkers and treatment efficacy, and the association between circulating tumor DNA (ctDNA) dynamics and PFS. Biomarker analysis revealed that the breast cancer gene 2, BMP/Retinoic Acid Inducible Neural Specific 3, F-box/WD repeat-containing protein 7, tyrosine-protein kinase KIT and retinoblastoma 1 abnormalities led to shorter PFS, while ctDNA negative at baseline or clearance at 2 cycles of treatment was associated with longer PFS (18.1 vs. 4.3 months). Taken together, sintilimab in combination with 2 cycles of nab-paclitaxel/carboplatin treatment produced encouraging PFS and better tolerability as first-line treatment for advanced sq-NSCLC.
Collapse
Affiliation(s)
- Mina Zhang
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University/Henan Cancer Hospital, 127 Dongming Rd, Zhengzhou, 450003, China
| | - Guowei Zhang
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University/Henan Cancer Hospital, 127 Dongming Rd, Zhengzhou, 450003, China
| | - Yuanyuan Niu
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University/Henan Cancer Hospital, 127 Dongming Rd, Zhengzhou, 450003, China
| | - Guifang Zhang
- Department of Medical Oncology, Xinxiang Central Hospital, 56 Jinsui Rd, Xinxiang, 453000, China
| | - Yinghua Ji
- Department of Medical Oncology, The First Affiliated Hospital of Xinxiang Medical University, 88 Jiankang Rd, Xinxiang, 453199, China
| | - Xiangtao Yan
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University/Henan Cancer Hospital, 127 Dongming Rd, Zhengzhou, 450003, China
| | - Xiaojuan Zhang
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University/Henan Cancer Hospital, 127 Dongming Rd, Zhengzhou, 450003, China
| | - Qichuan Wang
- Department of Medical Oncology, The Second People's Hospital of Nanyang, 66 Jianshe Rd, Nanyang, 473000, China
| | - Xiaohui Jing
- Department of Medical Oncology, The First People's Hospital of Pingdingshan, 117 Youyue Rd, Pingdingshan, 467099, China
| | - Junsheng Wang
- Department of Medical Oncology, Anyang Cancer Hospital, 2 N Huanbin Rd, Anyang, 455001, China
| | - Zhiyong Ma
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University/Henan Cancer Hospital, 127 Dongming Rd, Zhengzhou, 450003, China
| | - Huijuan Wang
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University/Henan Cancer Hospital, 127 Dongming Rd, Zhengzhou, 450003, China.
| |
Collapse
|
13
|
Pashkina E, Aktanova A, Boeva O, Bykova M, Gavrilova E, Goiman E, Kovalenko E, Saleh N, Grishina L, Kozlov V. Evaluation of the Immunosafety of Cucurbit[n]uril In Vivo. Pharmaceutics 2024; 16:127. [PMID: 38276497 PMCID: PMC10820314 DOI: 10.3390/pharmaceutics16010127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/08/2024] [Accepted: 01/16/2024] [Indexed: 01/27/2024] Open
Abstract
Cucurbiturils are a family of macrocyclic oligomers capable of forming host-guest complexes with various molecules. Due to noncovalent binding to drug molecules and low toxicity, cucurbiturils has been extensively investigated as potential carriers for drug delivery. However, the immune system's interactions with different drug carriers, including cucurbiturils, are still under investigation. In this study, we focused on cucurbiturils' immunosafety and immunomodulation properties in vivo. We measured blood counts and lymphocyte subpopulations in blood, spleen, and bone marrow, and assessed the in vivo toxicity to spleen and bone marrow cells after intraperitoneal administration to BALB/c mice. When assessing the effect of cucurbit[6]uril on blood parameters after three intraperitoneal injections within a week in laboratory animals, a decrease in white blood cells was found in mice after injections of cucurbit[6]util, but the observed decrease in the number of white blood cells was within the normal range. At the same time, cucurbit[7]uril and cucurbit[8]uril did not affect the leukocyte counts of mice after three injections. Changes in the number of platelets, erythrocytes, and monocytes, as well as in several other indicators, such as hematocrit or erythrocyte volumetric dispersion, were not detected. We show that cucurbiturils do not have immunotoxicity in vivo, with the exception of a cytotoxic effect on spleen cells after сucurbit[7]uril administration at a high dosage. We also evaluated the effect of cucurbiturils on cellular and humoral immune responses. We founded that cucurbiturils in high concentrations affect the immune system in vivo, and the action of various cucurbiturils differs in different homologues, which is apparently associated with different interactions in the internal environment of the body.
Collapse
Affiliation(s)
- Ekaterina Pashkina
- Research Institute of Fundamental and Clinical Immunology, 14, Yadrintsevskaya St., 630099 Novosibirsk, Russia
- Department of Clinical Immunology, Novosibirsk State Medical University, 52, Krasny Prospect, 630091 Novosibirsk, Russia
| | - Alina Aktanova
- Research Institute of Fundamental and Clinical Immunology, 14, Yadrintsevskaya St., 630099 Novosibirsk, Russia
- Department of Clinical Immunology, Novosibirsk State Medical University, 52, Krasny Prospect, 630091 Novosibirsk, Russia
| | - Olga Boeva
- Research Institute of Fundamental and Clinical Immunology, 14, Yadrintsevskaya St., 630099 Novosibirsk, Russia
| | - Maria Bykova
- Research Institute of Fundamental and Clinical Immunology, 14, Yadrintsevskaya St., 630099 Novosibirsk, Russia
| | - Elena Gavrilova
- Research Institute of Fundamental and Clinical Immunology, 14, Yadrintsevskaya St., 630099 Novosibirsk, Russia
| | - Elena Goiman
- Research Institute of Fundamental and Clinical Immunology, 14, Yadrintsevskaya St., 630099 Novosibirsk, Russia
| | | | - Na’il Saleh
- Department of Chemistry, College of Science, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates;
| | - Lyubov Grishina
- Research Institute of Fundamental and Clinical Immunology, 14, Yadrintsevskaya St., 630099 Novosibirsk, Russia
| | - Vladimir Kozlov
- Research Institute of Fundamental and Clinical Immunology, 14, Yadrintsevskaya St., 630099 Novosibirsk, Russia
| |
Collapse
|
14
|
Coschi CH, Juergens RA. Overcoming Resistance Mechanisms to Immune Checkpoint Inhibitors: Leveraging the Anti-Tumor Immune Response. Curr Oncol 2023; 31:1-23. [PMID: 38275827 PMCID: PMC10814017 DOI: 10.3390/curroncol31010001] [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: 10/25/2023] [Revised: 12/05/2023] [Accepted: 12/15/2023] [Indexed: 01/27/2024] Open
Abstract
As far back as 3000 years ago, the immune system was observed to play a role in mediating tumor regression. Since then, many strategies have been developed to leverage the anti-tumor immune response. However, while many patients respond to ICIs up front some do not, and many of those that do eventually experience tumor progression. Currently, there are several predictive biomarkers of the immune checkpoint inhibitor response; however, no one test appears to be universally predictive and their application varies by disease site. There are many ways in which cancer cells develop primary or acquired resistance to immune checkpoint inhibitors. Efforts to reverse resistance include ways to combat T cell exhaustion, reprogram the tumor microenvironment, increase the availability of tumor neo-antigens, target alternative immune checkpoints, restore a normal/healthy patient gut microbiome, oncolytic viruses and tumor vaccines. The most studied and most promising methods include combining ICIs with therapies targeting alternative immune checkpoints and restoring a normal/healthy patient gut microbiome. This review will discuss T cell-mediated immunity, how this is leveraged by modern immunotherapy to treat cancer and mechanisms of immune checkpoint inhibitor resistance, while highlighting strategies to overcome primary and secondary resistance mechanisms.
Collapse
Affiliation(s)
- Courtney H. Coschi
- Department of Oncology, McMaster University, 699 Concession Street, Hamilton, ON L8V 5C2, Canada;
| | - Rosalyn A. Juergens
- Department of Oncology, McMaster University, 699 Concession Street, Hamilton, ON L8V 5C2, Canada;
- Escarpment Cancer Research Institute, McMaster University, Hamilton, ON L8V 5C2, Canada
| |
Collapse
|
15
|
Lê H, Deforges J, Hua G, Idoux-Gillet Y, Ponté C, Lindner V, Olland A, Falcoz PE, Zaupa C, Jain S, Quéméneur E, Benkirane-Jessel N, Balloul JM. In vitro vascularized immunocompetent patient-derived model to test cancer therapies. iScience 2023; 26:108094. [PMID: 37860774 PMCID: PMC10582498 DOI: 10.1016/j.isci.2023.108094] [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: 06/15/2023] [Revised: 08/21/2023] [Accepted: 09/26/2023] [Indexed: 10/21/2023] Open
Abstract
This work describes a patient-derived tumoroid model (PDTs) to support precision medicine in lung oncology. The use of human adipose tissue-derived microvasculature and patient-derived peripheral blood mononuclear cells (PBMCs) permits to achieve a physiologically relevant tumor microenvironment. This study involved ten patients at various stages of tumor progression. The vascularized, immune-infiltrated PDT model could be obtained within two weeks, matching the requirements of the therapeutic decision. Histological and transcriptomic analyses confirmed that the main features from the original tumor were reproduced. The 3D tumor model could be used to determine the dynamics of response to antiangiogenic therapy and platinum-based chemotherapy. Antiangiogenic therapy showed a significant decrease in vascular endothelial growth factor (VEGF)-A expression, reflecting its therapeutic effect in the model. In an immune-infiltrated PDT model, chemotherapy showed the ability to decrease the levels of lymphocyte activation gene-3 protein (LAG-3), B and T lymphocyte attenuator (BTLA), and inhibitory receptors of T cells functions.
Collapse
Affiliation(s)
- Hélène Lê
- Transgene S.A, 400 Boulevard Gonthier d’Andernach, 67400 Illkirch-Graffenstaden, France
- INSERM UMR 1260, Regenerative Nanomedicine, 1 rue Eugène Boeckel, 67000 Strasbourg, France
| | - Jules Deforges
- Transgene S.A, 400 Boulevard Gonthier d’Andernach, 67400 Illkirch-Graffenstaden, France
| | - Guoqiang Hua
- INSERM UMR 1260, Regenerative Nanomedicine, 1 rue Eugène Boeckel, 67000 Strasbourg, France
| | - Ysia Idoux-Gillet
- INSERM UMR 1260, Regenerative Nanomedicine, 1 rue Eugène Boeckel, 67000 Strasbourg, France
| | - Charlotte Ponté
- INSERM UMR 1260, Regenerative Nanomedicine, 1 rue Eugène Boeckel, 67000 Strasbourg, France
- Hopitaux Universitaires de Strasbourg, 1 Place de l’Hôpital, 67000 Strasbourg, France
| | - Véronique Lindner
- INSERM UMR 1260, Regenerative Nanomedicine, 1 rue Eugène Boeckel, 67000 Strasbourg, France
| | - Anne Olland
- INSERM UMR 1260, Regenerative Nanomedicine, 1 rue Eugène Boeckel, 67000 Strasbourg, France
- Hopitaux Universitaires de Strasbourg, 1 Place de l’Hôpital, 67000 Strasbourg, France
| | - Pierre-Emanuel Falcoz
- INSERM UMR 1260, Regenerative Nanomedicine, 1 rue Eugène Boeckel, 67000 Strasbourg, France
- Hopitaux Universitaires de Strasbourg, 1 Place de l’Hôpital, 67000 Strasbourg, France
| | - Cécile Zaupa
- Boehringer Ingelheim, 29 avenue Tony Garnier, 69007 Lyon, France
| | - Shreyansh Jain
- Transgene S.A, 400 Boulevard Gonthier d’Andernach, 67400 Illkirch-Graffenstaden, France
| | - Eric Quéméneur
- Transgene S.A, 400 Boulevard Gonthier d’Andernach, 67400 Illkirch-Graffenstaden, France
| | - Nadia Benkirane-Jessel
- INSERM UMR 1260, Regenerative Nanomedicine, 1 rue Eugène Boeckel, 67000 Strasbourg, France
| | - Jean-Marc Balloul
- Transgene S.A, 400 Boulevard Gonthier d’Andernach, 67400 Illkirch-Graffenstaden, France
| |
Collapse
|
16
|
XIE FANGMEI, XI NAITE, HAN ZEPING, LUO WENFENG, SHEN JIAN, LUO JINGGENG, TANG XINGKUI, PANG TING, LV YUBING, LIANG JIABING, LIAO LIYIN, ZHANG HAOYU, JIANG YONG, LI YUGUANG, HE JINHUA. Progress in research on tumor microenvironment-based spatial omics technologies. Oncol Res 2023; 31:877-885. [PMID: 37744276 PMCID: PMC10513957 DOI: 10.32604/or.2023.029494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 06/21/2023] [Indexed: 09/26/2023] Open
Abstract
Spatial omics technology integrates the concept of space into omics research and retains the spatial information of tissues or organs while obtaining molecular information. It is characterized by the ability to visualize changes in molecular information and yields intuitive and vivid visual results. Spatial omics technologies include spatial transcriptomics, spatial proteomics, spatial metabolomics, and other technologies, the most widely used of which are spatial transcriptomics and spatial proteomics. The tumor microenvironment refers to the surrounding microenvironment in which tumor cells exist, including the surrounding blood vessels, immune cells, fibroblasts, bone marrow-derived inflammatory cells, various signaling molecules, and extracellular matrix. A key issue in modern tumor biology is the application of spatial omics to the study of the tumor microenvironment, which can reveal problems that conventional research techniques cannot, potentially leading to the development of novel therapeutic agents for cancer. This paper summarizes the progress of research on spatial transcriptomics and spatial proteomics technologies for characterizing the tumor immune microenvironment.
Collapse
Affiliation(s)
- FANGMEI XIE
- Central Laboratory, Panyu Central Hospital of Guangzhou, Guangzhou, China
| | - NAITE XI
- Central Laboratory, Panyu Central Hospital of Guangzhou, Guangzhou, China
| | - ZEPING HAN
- Central Laboratory, Panyu Central Hospital of Guangzhou, Guangzhou, China
| | - WENFENG LUO
- Central Laboratory, Panyu Central Hospital of Guangzhou, Guangzhou, China
| | - JIAN SHEN
- Central Laboratory, Panyu Central Hospital of Guangzhou, Guangzhou, China
| | - JINGGENG LUO
- Department of General Surgery, Panyu Central Hospital of Guangzhou, Guangzhou, China
| | - XINGKUI TANG
- Department of General Surgery, Panyu Central Hospital of Guangzhou, Guangzhou, China
| | - TING PANG
- Central Laboratory, Panyu Central Hospital of Guangzhou, Guangzhou, China
| | - YUBING LV
- Central Laboratory, Panyu Central Hospital of Guangzhou, Guangzhou, China
| | - JIABING LIANG
- Central Laboratory, Panyu Central Hospital of Guangzhou, Guangzhou, China
| | - LIYIN LIAO
- Central Laboratory, Panyu Central Hospital of Guangzhou, Guangzhou, China
| | - HAOYU ZHANG
- Central Laboratory, Panyu Central Hospital of Guangzhou, Guangzhou, China
| | - YONG JIANG
- Central Laboratory, Panyu Central Hospital of Guangzhou, Guangzhou, China
| | - YUGUANG LI
- Administrating Office, He Xian Memorial Hospital, Southern Medical University, Guangzhou, China
| | - JINHUA HE
- Central Laboratory, Panyu Central Hospital of Guangzhou, Guangzhou, China
| |
Collapse
|
17
|
Yan J, Jiang W, Kang G, Li Q, Tao L, Wang X, Yin J. Synergistic chemo-photo anticancer therapy by using reversible Diels-Alder dynamic covalent bond mediated polyprodrug amphiphiles and immunoactivation investigation. Biomater Sci 2023; 11:5819-5830. [PMID: 37439438 DOI: 10.1039/d3bm00889d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Highly efficient endocytosis and multi-approach integrated therapeutic tactics are important factors in oncotherapy. With the aid of thermally reversible furan-maleimide dynamic covalent bonds and the "polyprodrug amphiphiles" concept, thermo- and reduction-responsive PEG(-COOH)Fu/MI(-SS-)CPT copolymers were fabricated by the Diels-Alder (D-A) coupling of hydrophilic Fu(-COOH)-PEG and hydrophobic MI(-SS-)-CPT building blocks. The copolymers could self-assemble to form composite nanoparticles with a photothermal conversion reagent (IR780) and maintain excellent stability. In the in vitro simulated environments, the composite nanoparticles could detach Fu(-COOH)-PEG chains by a retro-D-A reaction upon near-infrared light (NIR) irradiation and reduce the size to facilitate endocytosis. Once in the intracellular environment, glutathione (GSH) could trigger a cascade reaction to release active CPT drugs to achieve chemotherapy, which could be further promoted by NIR light induced photothermal therapy. The in vivo mouse tumor model experiments demonstrated that these nanoparticles had an excellent therapeutic effect on solid tumors and inhibited their recurrence. Not only that, the synergistic chemical and optical therapy induced body immune response was also systematically evaluated; the maturation of dendritic cells, the proliferation of T cells, the increase of high mobility group box protein 1, and the decrease of immunosuppressive regulatory T cells confirmed that such synergistic therapy could effectively provide immune protection to the body. We believe such in situ generation of small-sized therapeutic units brought by a dynamically reversible D-A reaction could expand the pathway to design next generation drug delivery systems possessing superior design philosophy and excellent practice effects compared to currently available ones.
Collapse
Affiliation(s)
- Jinhao Yan
- Department of Polymer Science and Engineering, School of Chemistry and Chemical Engineering Hefei University of Technology and Anhui Province Key Laboratory of Advanced Catalytic Materials and Reaction Engineering Hefei, Anhui, 230009, P. R. China.
| | - Wenlong Jiang
- Department of Polymer Science and Engineering, School of Chemistry and Chemical Engineering Hefei University of Technology and Anhui Province Key Laboratory of Advanced Catalytic Materials and Reaction Engineering Hefei, Anhui, 230009, P. R. China.
| | - Guijie Kang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University Hefei, Anhui, 230032, P. R. China.
| | - Qingjie Li
- Department of Polymer Science and Engineering, School of Chemistry and Chemical Engineering Hefei University of Technology and Anhui Province Key Laboratory of Advanced Catalytic Materials and Reaction Engineering Hefei, Anhui, 230009, P. R. China.
| | - Longxiang Tao
- Department of Radiology, the First Affiliated Hospital of Anhui Medical University Hefei, Anhui, 230022, P. R. China.
| | - Xuefu Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University Hefei, Anhui, 230032, P. R. China.
| | - Jun Yin
- Department of Polymer Science and Engineering, School of Chemistry and Chemical Engineering Hefei University of Technology and Anhui Province Key Laboratory of Advanced Catalytic Materials and Reaction Engineering Hefei, Anhui, 230009, P. R. China.
| |
Collapse
|
18
|
Mamilos A, Lein A, Winter L, Ettl T, Künzel J, Reichert TE, Spanier G, Brochhausen C. Tumor Immune Microenvironment Heterogeneity at the Invasion Front and Tumor Center in Oral Squamous Cell Carcinoma as a Perspective of Managing This Cancer Entity. J Clin Med 2023; 12:jcm12041704. [PMID: 36836239 PMCID: PMC9958892 DOI: 10.3390/jcm12041704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/13/2023] [Accepted: 02/17/2023] [Indexed: 02/25/2023] Open
Abstract
BACKGROUND Evaluating the tumor microenvironment and its influence on clinical management and therapy response is becoming increasingly important. However, only a few studies deal with the spatial distribution of immune cells within the tumor. This study aimed to describe the topology of immune cells in the microenvironment of oral squamous cell carcinoma (OSCC) sectioned by tumor invasion front and tumor center and to test their prognostic relevance regarding patient survival. METHODS A total of 55 OSCC patient specimens were collected retrospectively. The cancer tissue was immunohistochemically stained using an automated tissue stainer Ventana Benchmark Ultra (Roche) and analyzed using discrete expression marker profiles on immune cells. We investigated CD4+ lymphocytes, CD8+ lymphocytes, CD68+ macrophages, CD163+ macrophages, and M1 macrophages regarding their spatial distribution. RESULTS The statistical analysis revealed that the quantity and distribution of CD4+ (p = 0.007), CD8+ (p < 0.001), CD68+ (p < 0.001), CD163+ cells (p = 0.004), and M1 (p < 0.001) macrophages were significantly higher at the invasion front compared to the tumor center in all observed cases. However, high and low immune cell counts in the tumor center and invasion front were not associated with overall survival. CONCLUSION Our results show two distinct immune microenvironments of the tumor center compared to the invasion front. Future studies are needed to explore how these results can be leveraged to improve patient therapy and outcome.
Collapse
Affiliation(s)
- Andreas Mamilos
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany
- Central Biobank Regensburg, University and University Hospital Regensburg, 93053 Regensburg, Germany
| | - Alexander Lein
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Lina Winter
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany
- Central Biobank Regensburg, University and University Hospital Regensburg, 93053 Regensburg, Germany
| | - Tobias Ettl
- Department of Oral and Maxillofacial Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Julian Künzel
- Department of Otorhinolaryngology, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Torsten E. Reichert
- Department of Oral and Maxillofacial Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Gerrit Spanier
- Department of Oral and Maxillofacial Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Christoph Brochhausen
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany
- Central Biobank Regensburg, University and University Hospital Regensburg, 93053 Regensburg, Germany
- Institute of Pathology, University Medical Centre Mannheim, Ruprecht-Karls-University of Heidelberg, 68167 Mannheim, Germany
- Correspondence: ; Tel.: +49-621-383-2275
| |
Collapse
|
19
|
Temozolomide combined with ipilimumab plus nivolumab enhances T cell killing of MGMT-expressing, MSS colorectal cancer cells. Am J Cancer Res 2023; 13:216-226. [PMID: 36777499 PMCID: PMC9906078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 12/09/2022] [Indexed: 02/14/2023] Open
Abstract
Colorectal cancer (CRC) is the third most frequently diagnosed cancer and third-deadliest cancer globally. Over 95% of patients with metastatic CRC have tumors that are microsatellite stable (MSS) and do not respond to immune checkpoint inhibitors (ICI). Results from the 2022 MAYA clinical trial suggest that the DNA-damaging agent temozolomide (TMZ), which is usually used to treat glioblastoma (GBM), sensitizes patients with MSS, MGMT-silenced CRC to ipilimumab + nivolumab ICI. The benefit of adding ipilimumab + nivolumab to TMZ and the impact of MGMT silencing remain unclear. Here, we aimed to determine in a controlled in vitro system if adding ICI to TMZ enhances T cell killing of MSS CRC cells. We also aimed to determine the contribution of MGMT to this response. Western blot analysis indicated that CRC cells (n = 4) had significantly elevated MGMT expression as compared to GBM cells (n = 4) likely due to MGMT promoter methylation in GBM cells. In line with this, CRC cells were slightly more resistant to TMZ compared to GBM cells after five days of treatment. TMZ + ICI sensitized MGMT-expressing, MSS CRC cells to T cell killing. TMZ alone did not enhance T cell killing of MSS or MSI CRC cells but did slightly enhance T cell killing of T98G GBM cells. Our results indicate that TMZ sensitizes MSS, MGMT-expressing CRC cells to ipilimumab + nivolumab ICI. Importantly, this suggests that TMZ-mediated sensitization to ipilimumab + nivolumab appears independent of MGMT status and the patient cohort that may benefit from TMZ + ipilimumab + nivolumab may be expanded to CRC patients with MGMT-expressing, MSS tumors.
Collapse
|
20
|
Song Z, Zou K, Zou L. Immune checkpoint blockade for locally advanced or recurrent/metastatic cervical cancer: An update on clinical data. Front Oncol 2022; 12:1045481. [PMID: 36644634 PMCID: PMC9832370 DOI: 10.3389/fonc.2022.1045481] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022] Open
Abstract
Immunotherapy has shown great promise in the field of oncology, and recent clinical trials have illustrated that immune checkpoint blockade (ICB) is safe and effective at treating a range of tumor types. Cervical cancer (CC) is the fourth most common malignancy in women. However, first-line treatments for locally advanced cervical cancer (LACC) and recurrent/metastatic (R/M) CC have limited efficacy. Thus, it is necessary to explore new treatment approaches. The National Comprehensive Cancer Network (NCCN) currently recommends pembrolizumab, a programmed cell death protein 1 (PD-1) monoclonal antibody, as a first line therapy for individuals with R/M CC. This study reviews the progress of ICB therapy for LACC and R/M CC and describes the current status of the combination of ICB therapy and other therapeutic modalities, including radiotherapy, chemotherapy, targeted therapy, and other immunotherapies. The focus is placed on studies published since 2018 with the aim of highlighting novel CC-specific immunotherapeutic approaches and treatment targets.
Collapse
Affiliation(s)
- Zhuo Song
- Department of Radiation Oncology, The Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, China
| | - Kun Zou
- Department of Radiation Oncology, The First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, China
| | - Lijuan Zou
- Department of Radiation Oncology, The Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|