1
|
Yu H, Zhu W, Lin C, Jia M, Tan X, Yuan Z, Feng S, Yan P. Stromal and tumor immune microenvironment reprogramming through multifunctional cisplatin-based liposomes boosts the efficacy of anti-PD-1 immunotherapy in pancreatic cancer. Biomater Sci 2023; 12:116-133. [PMID: 37921708 DOI: 10.1039/d3bm01118f] [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: 11/04/2023]
Abstract
The dense stromal barrier in pancreatic cancer tissues blocks intratumoral delivery and distribution of chemotherapeutics and therapeutic antibodies, causing poor chemoimmunotherapy responses. We designed a multi-targeted pH-sensitive liposome which encapsulates cisplatin (Pt) in its water core (denoted as ATF@Pt Lps) and shows high affinity for uPAR receptors in pancreatic cancer cells, tumor-associated macrophages, and cancer-associated fibroblasts. Systemic administration of ATF@Pt Lps enabled overcoming the central stromal cellular barrier and effective drug delivery into tumor cells, resulting in a strong therapeutic response in a Panc02 cell derived transplanted tumor mouse model. More importantly, ATF@Pt Lps degradation of collagen contributes to the infiltration of CD8+ T cells into tumors as well as an enhanced accumulation of anti PD-1 monoclonal antibodies. Furthermore, the killing of tumor cells by Pt also leads to the release of tumor antigens, which promote the proliferation of immune cells, especially CD83+ cells, Th1 CD4+ cells, and CD8+ cytotoxic T cells, that converted an immunoscore "cold" pancreatic cancer into a pro-immune "hot" tumor. A further combination with an immune checkpoint agent, anti PD-1 antibodies that inhibit PD-1, can enhance tumor specific cytotoxic T cell response. Accordingly, ATF@Pt Lps displays multi-targeting, controlled drug release, stromal disruption, enhanced penetration, killing of cancer cells, modification of the immunosuppressive microenvironment, and enhancement of immunity. This study provides important mechanistic information for the further development of a combination of ATF@Pt Lps and anti PD-1 antibodies for the effective treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Hang Yu
- Department of Pharmacy, Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangzhou, 510150, China.
| | - Wenting Zhu
- Department of Pharmacy, Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangzhou, 510150, China.
| | - Caiyan Lin
- Department of Pharmacy, Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangzhou, 510150, China.
| | - Menglei Jia
- Department of Pharmacy, Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangzhou, 510150, China.
| | - Xiaoxiao Tan
- Department of Pharmacy, Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangzhou, 510150, China.
| | - Zhongwen Yuan
- Department of Pharmacy, Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangzhou, 510150, China.
| | - Senglin Feng
- Department of Pharmacy, Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangzhou, 510150, China.
| | - Pengke Yan
- Department of Pharmacy, Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangzhou, 510150, China.
| |
Collapse
|
2
|
Bazsefidpar P, Eftekhar E, Jahromi MZ, Nikpoor AR, Moghadam ME, Zolghadri S. In-vitro cytotoxicity and in-vivo antitumor activity of two platinum complexes with 1,3-dimethyl pentyl glycine ligand against breast cancer. J Inorg Biochem 2023; 241:112144. [PMID: 36706492 DOI: 10.1016/j.jinorgbio.2023.112144] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 01/16/2023] [Accepted: 01/19/2023] [Indexed: 01/22/2023]
Abstract
Platinum (Pt) derivatives are good candidates for discovering new anti-tumor agents. The present research aims to explore the in-vivo and in-vitro anticancer activity of two platinum complexes with 1,3-dimethyl pentyl glycine ligand (DMPG), [Pt(bpy)(13DMPG)]NO3 and [Pt(dach)(13DMPG)]NO3, against breast cancer cells. The present study was conducted to investigate the cytotoxic potential of these compounds (2-400 μM) compared to standard drugs (cisplatin, oxaliplatin, and carboplatin) on SKBR3 cells using the methyl thiazol-tetrazolium (MTT) assay. Furthermore, the gene expression changes of Bak, Bim, Bcl-2, Caspase-3, and Caspase-9 were carried out by real-time polymerase chain reaction (PCR), and flow cytometric analysis was performed to confirm the cell apoptosis in the presence of the compounds. For more validation, in-vivo anticancer activities of both compounds were investigated against breast transplanted tumors in the BALB/c mice model. The cytotoxic studies by MTT assay revealed the anti-proliferative potential of both derivatives. [Pt(dach)(13DMPG)]NO3 with an IC50 value of 15 μM, exhibited higher cytotoxicity against SKBR3 cells as compared to [Pt(bpy)(13DMPG)]NO3, oxaliplatin, and carboplatin. Based on the flow cytometry analysis, both derivatives demonstrated apoptotic effects. Also, real-time PCR analysis revealed an up-regulation of Bak, Bim, Bax, Caspases-3, and Caspase-9 genes and a significant reduction in Bcl-2 gene expression in treated cells with both compounds compared to the control group. In-vivo results validated in-vitro analysis and showed the anticancer activity of compounds against breast transplanted tumors in the BALB/c mice model. According to the results, [Pt(dach)(13DMPG)]NO3 displayed a significant anticancer activity.
Collapse
Affiliation(s)
- Parisa Bazsefidpar
- Department of Biology, Jahrom Branch, Islamic Azad University, Jahrom, Iran
| | - Ebrahim Eftekhar
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | | | - Amin Reza Nikpoor
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | | | - Samaneh Zolghadri
- Department of Biology, Jahrom Branch, Islamic Azad University, Jahrom, Iran.
| |
Collapse
|
3
|
Patel M, Gbadegesin RA. Update on prognosis driven classification of pediatric AKI. Front Pediatr 2022; 10:1039024. [PMID: 36340722 PMCID: PMC9634036 DOI: 10.3389/fped.2022.1039024] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 10/03/2022] [Indexed: 11/29/2022] Open
Abstract
Acute kidney injury (AKI) affects a large proportion of hospitalized children and increases morbidity and mortality in this population. Initially thought to be a self-limiting condition with uniformly good prognosis, we now know that AKI can persist and progress to acute kidney disease (AKD) and chronic kidney disease (CKD). AKI is presently categorized by stage of injury defined by increase in creatinine, decrease in eGFR, or decrease in urine output. These commonly used biomarkers of acute kidney injury do not change until the injury is well established and are unable to detect early stage of the disease when intervention is likely to reverse injury. The kidneys have the ability to compensate and return serum creatinine to a normal or baseline level despite nephron loss in the setting of AKI possibly masking persistent dysfunction. Though these definitions are important, classifying children by their propensity for progression to AKD and CKD and defining these risk strata by other factors besides creatinine may allow for better prognosis driven discussion, expectation setting, and care for our patients. In order to develop a classification strategy, we must first be able to recognize children who are at risk for AKD and CKD based on modifiable and non-modifiable factors as well as early biomarkers that identify their risk of persistent injury. Prevention of initial injury, prompt evaluation and treatment if injury occurs, and mitigating further injury during the recovery period may be important factors in decreasing risk of AKD and CKD after AKI. This review will cover presently used definitions of AKI, AKD, and CKD, recent findings in epidemiology and risk factors for AKI to AKD to CKD progression, novel biomarkers for early identification of AKI and AKI that may progress to CKD and future directions for improving outcome in children with AKI.
Collapse
Affiliation(s)
- Mital Patel
- Department of Pediatrics, Division of Pediatric Nephrology, Duke University, Durham, NC, United State
| | | |
Collapse
|
4
|
Kozlowski L, Bielawska K, Zhymaila A, Malyszko J. Chronic Kidney Disease Prevalence in Patients with Colorectal Cancer Undergoing Surgery. Diagnostics (Basel) 2022; 12:2137. [PMID: 36140538 PMCID: PMC9497923 DOI: 10.3390/diagnostics12092137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 08/31/2022] [Accepted: 08/31/2022] [Indexed: 11/23/2022] Open
Abstract
Colorectal cancer (CRC) is a common and mortal disease. Chronic kidney disease (CKD) is the relatively common comorbidity among cancer patients affecting the available therapy and outcomes. However, data on prevalence of CKD in patients with CRC undergoing surgery is limited. The aim of the study was to evaluate the prevalence of CKD in a cohort of 560 consecutive patients with CRC undergoing surgical treatment with curative intent. Neoadjuvant therapy in a form of radiotherapy or radiochemotherapy was administered before the surgery in 67 patients and in 86 patients, respectively. Results: CKD was reported in 10%, diabetes in 25%, and hypertension in 60%, while anemia was reported in 47%. The patients with CKD were more likely to be older and anemic with higher serum CRP, which reflects a general inflammatory state. Relative to patients without this therapy, patients undergoing neoadjuvant radiochemotherapy were older, had significantly lower eGFR and albumin, and higher creatinine, aspartate aminotransferase and INR, before the surgery. All CKD patients, except two, were older than 65 years of age. Conclusions: In order to ensure the best possible outcomes, CKD should be diagnosed and treated appropriately in oncology patients to prevent complications, so they may continue their therapy with the least interruption or discontinuation of treatment.
Collapse
Affiliation(s)
- Leszek Kozlowski
- Oncological Surgery Department with Specialized Cancer Treatment Units, Maria Sklodowska-Curie Bialystok Oncology Centre, 15-027 Białystok, Poland
| | - Katarzyna Bielawska
- Oncological Surgery Department with Specialized Cancer Treatment Units, Maria Sklodowska-Curie Bialystok Oncology Centre, 15-027 Białystok, Poland
| | - Alena Zhymaila
- Department of Nephrology, Dialysis & Internal Diseases, The Medical University of Warsaw, 02-091 Warszawa, Poland
| | - Jolanta Malyszko
- Department of Nephrology, Dialysis & Internal Diseases, The Medical University of Warsaw, 02-091 Warszawa, Poland
| |
Collapse
|
5
|
Cui Z, Ruan Z, Zeng J, Sun J, Ye W, Xu W, Guo X, Zhang L, Song L. Lung‐specific exosomes for co‐delivery of
CD47
blockade and cisplatin for the treatment of non–small cell lung cancer. Thorac Cancer 2022; 13:2723-2731. [PMID: 36054073 PMCID: PMC9527158 DOI: 10.1111/1759-7714.14606] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 07/22/2022] [Accepted: 07/26/2022] [Indexed: 12/25/2022] Open
Abstract
A cluster of differentiation 47 (CD47) and immune‐modulatory protein for myeloid cells has been implicated in cisplatin (CDDP) resistance. Exosome delivery of drugs has shown great potential for targeted drug delivery in the treatment of various diseases. In the current study, we explored the approach of co‐delivering CDDP and CD47 antibody with MDA‐MB‐231 cell‐derived exosome 231‐exo (CaCE) and assessed the phagocytosis activity of bone marrow flow cytometry derived macrophages (BMDM) against co‐cultured A549 cells. CD8+ T‐cell proliferation was examined with flow cytometry analysis. In vivo, we used the Lewis lung carcinoma (LLC) tumor‐bearing mouse model and assessed survival rate, tumor weight, phagocytosis, and T‐cell proliferation, as well as cytokine levels in tumors analyzed by enzyme‐linked immunoassay (ELISA). Although co‐administration of CDDP with anti‐CD47 (CDDP and aCD47) showed a significant antitumor effect, CaCE had an even more dramatic anticancer effect in survival rate and tumor weight. We observed increased phagocytosis activity selectively against lung tumor cells in vivo and in vitro with exosome CaCE treatment. CaCE treatment also increased T‐cell proliferation compared to the vehicle treatment and co‐administration groups. Furthermore, immunostimulatory interleukin (IL)‐12p and interferon (IFN)‐γ were increased, whereas transforming growth factor β (TGF‐β) were decreased, indicating the improved CDDP anticancer effect is related to a tumor microenvironmental change. Our study demonstrates a dramatically improved anticancer effect of CDDP when administered by exosome co‐delivery with anti‐CD47.
Collapse
Affiliation(s)
- Zhilei Cui
- Department of Respiratory Medicine XinHua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Zhengshang Ruan
- Department of Infectious Disease XinHua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Junxiang Zeng
- Department of Laboratory Medicine XinHua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Jinyuan Sun
- Department of Respiratory Medicine XinHua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Wenjing Ye
- Department of Respiratory Medicine XinHua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Weiguo Xu
- Department of Respiratory Medicine XinHua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Xuejun Guo
- Department of Respiratory Medicine XinHua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Linlin Zhang
- Department of Nuclear Medicine Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Lin Song
- Department of Respiratory Medicine XinHua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai China
| |
Collapse
|
6
|
Klumpers MJ, Witte WD, Gattuso G, Schiavello E, Terenziani M, Massimino M, Gidding CEM, Vermeulen SH, Driessen CM, van Herpen CM, van Meerten E, Guchelaar HJ, Coenen MJH, te Loo DMWM. Genome-Wide Analyses of Nephrotoxicity in Platinum-Treated Cancer Patients Identify Association with Genetic Variant in RBMS3 and Acute Kidney Injury. J Pers Med 2022; 12:jpm12060892. [PMID: 35743677 PMCID: PMC9224783 DOI: 10.3390/jpm12060892] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 05/25/2022] [Indexed: 02/06/2023] Open
Abstract
Nephrotoxicity is a common and dose-limiting side effect of platinum compounds, which often manifests as acute kidney injury or hypomagnesemia. This study aimed to investigate the genetic risk loci for platinum-induced nephrotoxicity. Platinum-treated brain tumor and head–neck tumor patients were genotyped with genome-wide coverage. The data regarding the patient and treatment characteristics and the laboratory results reflecting the nephrotoxicity during and after the platinum treatment were collected from the medical records. Linear and logistic regression analyses were performed to investigate the associations between the genetic variants and the acute kidney injury and hypomagnesemia phenotypes. A cohort of 195 platinum-treated patients was included, and 9,799,032 DNA variants passed the quality control. An association was identified between RBMS3 rs10663797 and acute kidney injury (coefficient −0.10 (95% confidence interval −0.13–−0.06), p-value 2.72 × 10−8). The patients who carried an AC deletion at this locus had statistically significantly lower glomerular filtration rates after platinum treatment. Previously reported associations, such as BACH2 rs4388268, could not be replicated in this study’s cohort. No statistically significant associations were identified for platinum-induced hypomagnesemia. The genetic variant in RBMS3 was not previously linked to nephrotoxicity or related traits. The validation of this study’s results in independent cohorts is needed to confirm this novel association.
Collapse
Affiliation(s)
- Marije J. Klumpers
- Department of Pediatrics, Radboud University Medical Center, Postbox 9101, 6500 HB Nijmegen, The Netherlands;
| | - Ward De Witte
- Department of Human Genetics, Radboud University Medical Center, Postbox 9101, 6500 HB Nijmegen, The Netherlands; (W.D.W.); (M.J.H.C.)
| | - Giovanna Gattuso
- Pediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian, 1, 20133 Milan, Italy; (G.G.); (E.S.); (M.T.); (M.M.)
| | - Elisabetta Schiavello
- Pediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian, 1, 20133 Milan, Italy; (G.G.); (E.S.); (M.T.); (M.M.)
| | - Monica Terenziani
- Pediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian, 1, 20133 Milan, Italy; (G.G.); (E.S.); (M.T.); (M.M.)
| | - Maura Massimino
- Pediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian, 1, 20133 Milan, Italy; (G.G.); (E.S.); (M.T.); (M.M.)
| | - Corrie E. M. Gidding
- Princess Maxima Center for Pediatric Oncology, Postbox 113, 3720 AC Bilthoven, The Netherlands;
| | - Sita H. Vermeulen
- Department for Health Evidence, Radboud University Medical Center, Postbox 9101, 6500 HB Nijmegen, The Netherlands;
| | - Chantal M. Driessen
- Department of Medical Oncology, Radboud University Medical Center, Postbox 9101, 6500 HB Nijmegen, The Netherlands; (C.M.D.); (C.M.v.H.)
| | - Carla M. van Herpen
- Department of Medical Oncology, Radboud University Medical Center, Postbox 9101, 6500 HB Nijmegen, The Netherlands; (C.M.D.); (C.M.v.H.)
| | - Esther van Meerten
- Department of Medical Oncology, Erasmus MC Cancer Institute, Postbox 2040, 3000 CA Rotterdam, The Netherlands;
| | - Henk-Jan Guchelaar
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, Postbox 9600, 2300 RC Leiden, The Netherlands;
| | - Marieke J. H. Coenen
- Department of Human Genetics, Radboud University Medical Center, Postbox 9101, 6500 HB Nijmegen, The Netherlands; (W.D.W.); (M.J.H.C.)
| | - D. Maroeska W. M. te Loo
- Department of Pediatrics, Radboud University Medical Center, Postbox 9101, 6500 HB Nijmegen, The Netherlands;
- Correspondence: ; Tel.: +31-24-361-44-15
| |
Collapse
|