1
|
Pienkowski T, Wawrzak-Pienkowska K, Tankiewicz-Kwedlo A, Ciborowski M, Kurek K, Pawlak D. Leveraging glycosylation for early detection and therapeutic target discovery in pancreatic cancer. Cell Death Dis 2025; 16:227. [PMID: 40164585 PMCID: PMC11958638 DOI: 10.1038/s41419-025-07517-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: 11/18/2024] [Revised: 02/17/2025] [Accepted: 03/11/2025] [Indexed: 04/02/2025]
Abstract
Pancreatic cancer (PC) remains one of the most lethal malignancies, primarily due to late-stage diagnosis, limited biomarker specificity, and aggressive metastatic potential. Recent glycoproteomic studies have illuminated the crucial role of glycosylation in PC progression, revealing altered glycosylation patterns that impact cell adhesion, immune evasion, and tumor invasiveness. Biomarkers such as CA19-9 remain the clinical standard, yet limitations in sensitivity and specificity, especially in early disease stages, necessitate the exploration of alternative markers. Emerging glycoproteins-such as mesothelin, thrombospondin-2, and glycan modifications like sialyl-Lewis x-offer diagnostic promise when combined with CA19-9 or used in profiling panels. Furthermore, therapeutic strategies targeting glycosylation processes, including sialylation, and fucosylation, have shown potential in curbing PC metastasis and enhancing immune response. Translational platforms, such as patient-derived xenografts and advanced in vitro models, are pivotal in validating these findings and assessing glycosylation potential therapeutic impact. Continued exploration of glycosylation-driven mechanisms and biomarker discovery in PC can significantly advance early detection and treatment efficacy, offering new hope in the management of this challenging disease.
Collapse
Affiliation(s)
- Tomasz Pienkowski
- Clinical Research Center, Medical University of Bialystok, Sklodowskiej MC 24A, Bialystok, Poland
- Department of Pharmacodynamics, Medical University of Bialystok, Bialystok, Poland
| | - Katarzyna Wawrzak-Pienkowska
- Department of Gastroenterology and Internal Medicine, Medical University of Bialystok, Bialystok, Poland
- Department of Gastroenterology, Hepatology and Internal Diseases, Voivodeship Hospital in Bialystok, Bialystok, Poland
| | | | - Michal Ciborowski
- Clinical Research Center, Medical University of Bialystok, Sklodowskiej MC 24A, Bialystok, Poland
| | - Krzysztof Kurek
- Department of Gastroenterology and Internal Medicine, Medical University of Bialystok, Bialystok, Poland
| | - Dariusz Pawlak
- Department of Pharmacodynamics, Medical University of Bialystok, Bialystok, Poland.
| |
Collapse
|
2
|
Kapare H, Bhosale M, Bhole R. Navigating the future: Advancements in monoclonal antibody nanoparticle therapy for cancer. J Drug Deliv Sci Technol 2025; 104:106495. [DOI: 10.1016/j.jddst.2024.106495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
3
|
Barros AG, Mansinho H, Couto N, Teixeira MR, Tonin FS, Francisco R, Duarte-Ramos F. The Initial Journey of Patients with Metastatic Pancreatic Cancer (PaCTO Project): A Nationwide Survey among Portuguese Specialist Physicians. GE PORTUGUESE JOURNAL OF GASTROENTEROLOGY 2024; 31:262-272. [PMID: 39114325 PMCID: PMC11305690 DOI: 10.1159/000533178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 07/18/2023] [Indexed: 08/10/2024]
Abstract
Introduction We aimed to characterize the initial healthcare journey of metastatic pancreatic ductal adenocarcinoma (mPDAC) patients in Portugal, including healthcare provision and factors affecting therapeutic decisions, namely BRCA mutations testing. Methods This is a descriptive cross-sectional, web-based survey using a convenience sampling approach. Portuguese oncologists and pathologists that routinely work with mPDAC patients from the different geographical regions and settings were invited to participate in the study via email (December 2020). Descriptive statistical analyses were performed, with categorical variables reported as absolute and relative frequencies, and continuous variables with non-normal distribution as median and interquartile range (IQR) (Stata v.15.0). Results Seventy physicians participated in the study (43 oncologists, 27 pathologists). According to the responses, a median of 28 patients per center (IQR 12-70) was diagnosed with PDAC in the previous year; 22 of them referring (IQR 8-70) to mPDAC. The pointed median time from patients' first hospital admission until disease diagnosis/staging is between 2 and 4 weeks. Endoscopic ultrasound with fine-needle biopsy is available in most hospitals (86%). Around 50% of physicians request BRCA testing; the assessment of additional biomarkers besides BRCA is requested by 40% of professionals. Half of them stated that BRCA testing should be requested earlier-upon histological diagnosis, especially because the median time for results is of 4.0 weeks (IQR 4-8). PARP inhibitors such as olaparib, when available, would be the therapy of choice for most oncologists (71%) if no disease' progression occurs after 4 months. Treatments' selection is usually grounded on clinical criteria (e.g., performance status, liver function). Around 45% of patients use FOLFIRINOX/mFOLFIRINOX as the first-line therapy. Gemcitabine + nab-paclitaxel is used by 35% of patients as the second-line therapy. Conclusions Physicians in Portugal support the increasing role of patient-tailored treatments in mPDAC, whose selection should be grounded on tumoral subtyping and molecular profiling. Further efforts to develop multidisciplinary teams, standardized clinical practice, and optimize the implementation of new target therapies are needed.
Collapse
Affiliation(s)
- Anabela G. Barros
- Department of Medical Oncology, University Hospital of Coimbra, Coimbra, Portugal
| | - Hélder Mansinho
- Hemato Oncology Department, Garcia de Orta Hospital, Almada, Portugal
| | - Nuno Couto
- Digestive Unit, Champalimaud Clinical Centre, Champalimaud Research Centre, Lisbon, Portugal
| | - Manuel R. Teixeira
- Department of Laboratory Genetics, Portuguese Oncology Institute of Porto (IPO Porto), Porto Comprehensive Cancer Center, Porto, Portugal
- Cancer Genetics Group, IPO Porto Research Center (CI-IPOP)/RISE@CI-IPOP (Health Research Network), School of Medicine and Biomedical Sciences (ICBAS), University of Porto, Porto, Portugal
| | - Fernanda S. Tonin
- Pharmaceutical Sciences Postgraduate Program, Federal University of Parana, Curitiba, Brazil
- H&TRC–Health & Technology Research Center, ESTeSL- Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, Lisbon, Portugal
| | | | - Filipa Duarte-Ramos
- Department of Pharmacy, Pharmacology and Health Technologies, Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
- Research Institute for Medicine (iMed.ULisboa), Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
- EPIUnit, Instituto de Saúde Pública da Universidade do Porto (ISPUP), Laboratório para a Investigação Integrativa e Translacional em Saúde Populacional (ITR), Porto, Portugal
| |
Collapse
|
4
|
Ma Z, Zhou Z, Duan W, Yao G, Sheng S, Zong S, Zhang X, Li C, Liu Y, Ou F, Dahar MR, Huang Y, Yu L. DR30318, a novel tri-specific T cell engager for Claudin 18.2 positive cancers immunotherapy. Cancer Immunol Immunother 2024; 73:82. [PMID: 38554200 PMCID: PMC10981630 DOI: 10.1007/s00262-024-03673-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: 02/20/2024] [Accepted: 03/08/2024] [Indexed: 04/01/2024]
Abstract
BACKGROUND Claudin 18.2 (CLDN18.2) is a highly anticipated target for solid tumor therapy, especially in advanced gastric carcinoma and pancreatic carcinoma. The T cell engager targeting CLDN18.2 represents a compelling strategy for enhancing anti-cancer efficacy. METHODS Based on the in-house screened anti-CLDN18.2 VHH, we have developed a novel tri-specific T cell engager targeting CLDN18.2 for gastric and pancreatic cancer immunotherapy. This tri-specific antibody was designed with binding to CLDN18.2, human serum albumin (HSA) and CD3 on T cells. RESULTS The DR30318 demonstrated binding affinity to CLDN18.2, HSA and CD3, and exhibited T cell-dependent cellular cytotoxicity (TDCC) activity in vitro. Pharmacokinetic analysis revealed a half-life of 22.2-28.6 h in rodents and 41.8 h in cynomolgus monkeys, respectively. The administration of DR30318 resulted in a slight increase in the levels of IL-6 and C-reactive protein (CRP) in cynomolgus monkeys. Furthermore, after incubation with human PBMCs and CLDN18.2 expressing cells, DR30318 induced TDCC activity and the production of interleukin-6 (IL-6) and interferon-gamma (IFN-γ). Notably, DR30318 demonstrated significant tumor suppression effects on gastric cancer xenograft models NUGC4/hCLDN18.2 and pancreatic cancer xenograft model BxPC3/hCLDN18.2 without affecting the body weight of mice.
Collapse
Affiliation(s)
- Zhe Ma
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang Province, China
- Department of Innovative Drug Discovery and Development, Zhejiang Doer Biologics Co., Ltd., Hangzhou, 310058, Zhejiang Province, China
| | - Zhenxing Zhou
- Department of Innovative Drug Discovery and Development, Zhejiang Doer Biologics Co., Ltd., Hangzhou, 310058, Zhejiang Province, China
| | - Wenwen Duan
- Department of Innovative Drug Discovery and Development, Zhejiang Doer Biologics Co., Ltd., Hangzhou, 310058, Zhejiang Province, China
| | - Gaofeng Yao
- Department of Innovative Drug Discovery and Development, Zhejiang Doer Biologics Co., Ltd., Hangzhou, 310058, Zhejiang Province, China
| | - Shimei Sheng
- Department of Innovative Drug Discovery and Development, Zhejiang Doer Biologics Co., Ltd., Hangzhou, 310058, Zhejiang Province, China
| | - Sidou Zong
- Department of Innovative Drug Discovery and Development, Zhejiang Doer Biologics Co., Ltd., Hangzhou, 310058, Zhejiang Province, China
| | - Xin Zhang
- Department of Innovative Drug Discovery and Development, Zhejiang Doer Biologics Co., Ltd., Hangzhou, 310058, Zhejiang Province, China
| | - Changkui Li
- Department of Innovative Drug Discovery and Development, Zhejiang Doer Biologics Co., Ltd., Hangzhou, 310058, Zhejiang Province, China
| | - Yuanyuan Liu
- Department of Innovative Drug Discovery and Development, Zhejiang Doer Biologics Co., Ltd., Hangzhou, 310058, Zhejiang Province, China
| | - Fengting Ou
- Jinhua Institute of Zhejiang University, Jinhua, 321036, China
| | - Maha Raja Dahar
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang Province, China
| | - Yanshan Huang
- Department of Innovative Drug Discovery and Development, Zhejiang Doer Biologics Co., Ltd., Hangzhou, 310058, Zhejiang Province, China.
| | - Lushan Yu
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang Province, China.
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, 310058, China.
- Department of Pharmacy, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China.
- Jinhua Institute of Zhejiang University, Jinhua, 321036, China.
- Department of Pharmacy, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, 312000, China.
| |
Collapse
|
5
|
Gupta MK, Vadde R. Delivery strategies of immunotherapies in the treatment of pancreatic cancer. IMMUNE LANDSCAPE OF PANCREATIC CANCER DEVELOPMENT AND DRUG RESISTANCE 2024:173-202. [DOI: 10.1016/b978-0-443-23523-8.00004-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
6
|
Busato D, Capolla S, Durigutto P, Mossenta M, Bozzer S, Sblattero D, Macor P, Dal Bo M, Toffoli G. A novel complement-fixing IgM antibody targeting GPC1 as a useful immunotherapeutic strategy for the treatment of pancreatic ductal adenocarcinoma. J Transl Med 2023; 21:864. [PMID: 38017492 PMCID: PMC10685509 DOI: 10.1186/s12967-023-04745-9] [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: 10/06/2023] [Accepted: 11/20/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive cancers with a very low survival rate at 5 years. The use of chemotherapeutic agents results in only modest prolongation of survival and is generally associated with the occurrence of toxicity effects. Antibody-based immunotherapy has been proposed for the treatment of PDAC, but its efficacy has so far proved limited. The proteoglycan glypican-1 (GPC1) may be a useful immunotherapeutic target because it is highly expressed on the surface of PDAC cells, whereas it is not expressed or is expressed at very low levels in benign neoplastic lesions, chronic pancreatitis, and normal adult tissues. Here, we developed and characterized a specific mouse IgM antibody (AT101) targeting GPC1. METHODS We developed a mouse monoclonal antibody of the IgM class directed against an epitope of GPC1 in close proximity to the cell membrane. For this purpose, a 46 amino acid long peptide of the C-terminal region was used to immunize mice by an in-vivo electroporation protocol followed by serum titer and hybridoma formation. RESULTS The ability of AT101 to bind the GPC1 protein was demonstrated by ELISA, and by flow cytometry and immunofluorescence analysis in the GPC1-expressing "PDAC-like" BXPC3 cell line. In-vivo experiments in the BXPC3 xenograft model showed that AT101 was able to bind GPC1 on the cell surface and accumulate in the BXPC3 tumor masses. Ex-vivo analyses of BXPC3 tumor masses showed that AT101 was able to recruit immunological effectors (complement system components, NK cells, macrophages) to the tumor site and damage PDAC tumor tissue. In-vivo treatment with AT101 reduced tumor growth and prolonged survival of mice with BXPC3 tumor (p < 0.0001). CONCLUSIONS These results indicate that AT101, an IgM specific for an epitope of GPC1 close to PDAC cell surface, is a promising immunotherapeutic agent for GPC1-expressing PDAC, being able to selectively activate the complement system and recruit effector cells in the tumor microenvironment, thus allowing to reduce tumor mass growth and improve survival in treated mice.
Collapse
Affiliation(s)
- Davide Busato
- Experimental and Clinical Pharmacology, Centro Di Riferimento Oncologico (CRO) Di Aviano IRCCS, 33081, Aviano, Italy
- Department of Life Sciences, University of Trieste, 34127, Trieste, Italy
| | - Sara Capolla
- Experimental and Clinical Pharmacology, Centro Di Riferimento Oncologico (CRO) Di Aviano IRCCS, 33081, Aviano, Italy
| | - Paolo Durigutto
- Department of Life Sciences, University of Trieste, 34127, Trieste, Italy
| | - Monica Mossenta
- Experimental and Clinical Pharmacology, Centro Di Riferimento Oncologico (CRO) Di Aviano IRCCS, 33081, Aviano, Italy
- Department of Life Sciences, University of Trieste, 34127, Trieste, Italy
| | - Sara Bozzer
- Experimental and Clinical Pharmacology, Centro Di Riferimento Oncologico (CRO) Di Aviano IRCCS, 33081, Aviano, Italy
| | - Daniele Sblattero
- Department of Life Sciences, University of Trieste, 34127, Trieste, Italy
| | - Paolo Macor
- Department of Life Sciences, University of Trieste, 34127, Trieste, Italy
| | - Michele Dal Bo
- Experimental and Clinical Pharmacology, Centro Di Riferimento Oncologico (CRO) Di Aviano IRCCS, 33081, Aviano, Italy.
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology, Centro Di Riferimento Oncologico (CRO) Di Aviano IRCCS, 33081, Aviano, Italy
| |
Collapse
|
7
|
Sun J, Baker JR, Russell CC, Pham HNT, Goldsmith CD, Cossar PJ, Sakoff JA, Scarlett CJ, McCluskey A. Novel piperazine-1,2,3-triazole leads for the potential treatment of pancreatic cancer. RSC Med Chem 2023; 14:2246-2267. [PMID: 37974967 PMCID: PMC10650957 DOI: 10.1039/d2md00289b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 06/26/2023] [Indexed: 11/19/2023] Open
Abstract
From lead 1, (N-(4-((4-(3-(4-(3-methoxyphenyl)-1H-1,2,3-triazol-1-yl)propyl)piperazin-1-yl)sulfonyl)-phenyl)acetamide), a S100A2-p53 protein-protein interaction inhibitor based on an in silico modelling driven hypothesis, four focused libraries were designed and synthesised. Growth inhibition screening was performed against 16 human cancer cell lines including the pancreatic cell lines MiaPaCa2, BxPC3, AsPC-1, Capan-2, HPAC, PANC-1 and the drug resistant CFPAC1. Modification of 1's phenylacetamide moiety, gave Library 1 with only modest pancreatic cancer activity. Modification of the 3-OCH3Ph moiety (Library 2) gave 4-CH3 (26), 4-CH2CH3 (27), 4-CF3 (31) and 4-NO2 (32) with sterically bulky groups more active. A 4-CF3 acetamide replacement enhanced cytotoxicity (Library 3). The 4-C(CH3)336 resulted in a predicted steric clash in the S100A2-p53 binding groove, with a potency decrease. Alkyl moieties afforded more potent analogues, 34 (4-CH3) and 35 (CH2CH3), a trend evident against pancreatic cancer: GI50 3.7 (35; BxPC-3) to 18 (40; AsPC-1) μM. Library 4 analogues with a 2-CF3 and 3-CF3 benzenesulfonamide moiety were less active than the corresponding Library 3 analogues. Two additional analogues were designed: 51 (4-CF3; 4-OCH3) and 52 (4-CF3; 2-OCH3) revealed 52 to be 10-20 fold more active than 51, against the pancreatic cancer cell lines examined with sub-micromolar GI50 values 0.43 (HPAC) to 0.61 μM (PANC-1). MOE calculated binding scores for each pose are also consistent with the observed biological activity with 52. The obtained SAR data is consistent with the proposed interaction within the S100A2-p53 bonding groove.
Collapse
Affiliation(s)
- Jufeng Sun
- Chemistry, School of Environmental & Life Sciences, The University of Newcastle University Drive Callaghan NSW 2308 Australia
- Medicinal Chemistry, School of Pharmacy, Binzhou Medical University Yantai 264003 China
| | - Jennifer R Baker
- Chemistry, School of Environmental & Life Sciences, The University of Newcastle University Drive Callaghan NSW 2308 Australia
| | - Cecilia C Russell
- Chemistry, School of Environmental & Life Sciences, The University of Newcastle University Drive Callaghan NSW 2308 Australia
| | - Hong N T Pham
- Experimental Therapeutics Group, Department of Medical Oncology, Calvary Mater Newcastle Hospital Edith Street Waratah NSW 2298 Australia
| | - Chloe D Goldsmith
- Experimental Therapeutics Group, Department of Medical Oncology, Calvary Mater Newcastle Hospital Edith Street Waratah NSW 2298 Australia
| | - Peter J Cossar
- Chemistry, School of Environmental & Life Sciences, The University of Newcastle University Drive Callaghan NSW 2308 Australia
| | - Jennette A Sakoff
- Experimental Therapeutics Group, Department of Medical Oncology, Calvary Mater Newcastle Hospital Edith Street Waratah NSW 2298 Australia
| | - Christopher J Scarlett
- School of Environmental & Life Sciences, The University of Newcastle Ourimbah NSW 2258 Australia
| | - Adam McCluskey
- Chemistry, School of Environmental & Life Sciences, The University of Newcastle University Drive Callaghan NSW 2308 Australia
| |
Collapse
|
8
|
Singh M, Jana BK, Pal P, Singha I, Rajkumari A, Chowrasia P, Nath V, Mazumder B. Nanoparticles in pancreatic cancer therapy: a detailed and elaborated review on patent literature. Expert Opin Ther Pat 2023; 33:681-699. [PMID: 37991186 DOI: 10.1080/13543776.2023.2287520] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 11/21/2023] [Indexed: 11/23/2023]
Abstract
INTRODUCTION Nanotechnology may open up new avenues for overcoming the challenges of pancreatic cancer therapy as a broad arsenal of anticancer medicines fail to realize their full therapeutic potential in pancreatic ductal adenocarcinoma due to the formation of multiple resistance mechanisms inside the tumor. Many studies have reported the successful use of various nano formulations in pancreatic cancer therapy. AREAS COVERED This review covers all the major nanotechnology-based patent litrature available on renowned patent data bases like Patentscope and Espacenet, through the time period of 2007-2022. This is an entirely patent centric review, and it includes both clinical and non-clinical data available on nanotechnology-based therapeutics and diagnostic tools for pancreatic cancer. EXPERT OPINION For the sake of understanding, the patents are categorized under various formulation-specific heads like metallic/non-metallic nanoparticles, polymeric nanoparticles, liposomes, carbon nanotubes, protein nanoparticles and liposomes. This distinguishes one specific nanoparticle type from another and makes this review a one-of-a-kind comprehensive patent compilation that has not been reported so far in the history of nanotechnological formulations in pancreatic cancer.
Collapse
Affiliation(s)
- Mohini Singh
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, India
| | - Bani Kumar Jana
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, India
| | - Paulami Pal
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, India
| | - Ishita Singha
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, India
| | - Ananya Rajkumari
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, India
| | - Pinky Chowrasia
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, India
| | - Venessa Nath
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, India
| | - Bhaskar Mazumder
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, India
| |
Collapse
|
9
|
Olajubutu O, Ogundipe OD, Adebayo A, Adesina SK. Drug Delivery Strategies for the Treatment of Pancreatic Cancer. Pharmaceutics 2023; 15:pharmaceutics15051318. [PMID: 37242560 DOI: 10.3390/pharmaceutics15051318] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/15/2023] [Accepted: 04/19/2023] [Indexed: 05/28/2023] Open
Abstract
Pancreatic cancer is fast becoming a global menace and it is projected to be the second leading cause of cancer-related death by 2030. Pancreatic adenocarcinomas, which develop in the pancreas' exocrine region, are the predominant type of pancreatic cancer, representing about 95% of total pancreatic tumors. The malignancy progresses asymptomatically, making early diagnosis difficult. It is characterized by excessive production of fibrotic stroma known as desmoplasia, which aids tumor growth and metastatic spread by remodeling the extracellular matrix and releasing tumor growth factors. For decades, immense efforts have been harnessed toward developing more effective drug delivery systems for pancreatic cancer treatment leveraging nanotechnology, immunotherapy, drug conjugates, and combinations of these approaches. However, despite the reported preclinical success of these approaches, no substantial progress has been made clinically and the prognosis for pancreatic cancer is worsening. This review provides insights into challenges associated with the delivery of therapeutics for pancreatic cancer treatment and discusses drug delivery strategies to minimize adverse effects associated with current chemotherapy options and to improve the efficiency of drug treatment.
Collapse
Affiliation(s)
| | - Omotola D Ogundipe
- Department of Pharmaceutical Sciences, Howard University, Washington, DC 20059, USA
| | - Amusa Adebayo
- Department of Pharmaceutical Sciences, Howard University, Washington, DC 20059, USA
| | - Simeon K Adesina
- Department of Pharmaceutical Sciences, Howard University, Washington, DC 20059, USA
| |
Collapse
|
10
|
Dhanisha SS, Guruvayoorappan C. Pathological Implications of Mucin Signaling in Metastasis. Curr Cancer Drug Targets 2023; 23:585-602. [PMID: 36941808 DOI: 10.2174/1568009623666230320121332] [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: 06/10/2022] [Revised: 01/11/2023] [Accepted: 01/25/2023] [Indexed: 03/23/2023]
Abstract
The dynamic mucosal layer provides a selective protective barrier for the epithelial cells lining the body cavities. Diverse human malignancies exploit their intrinsic role to protect and repair epithelia for promoting growth and survival. Aberrant expression of mucin has been known to be associated with poor prognosis of many cancers. However, the emergence of new paradigms in the study of metastasis recognizes the involvement of MUC1, MUC4, MUC5AC, MUC5B, and MUC16 during metastasis initiation and progression. Hence mucins can be used as an attractive target in future diagnostic and therapeutic strategies. In this review, we discuss in detail about mucin family and its domains and the role of different mucins in regulating cancer progression and metastasis. In addition, we briefly discuss insights into mucins as a therapeutic agent.
Collapse
Affiliation(s)
| | - Chandrasekharan Guruvayoorappan
- Laboratory of Immunopharmacology and Experimental Therapeutics, Division of Cancer Research, Regional Cancer Centre, Medical College Campus, University of Kerala, Thiruvananthapuram, Kerala, 695011, India
| |
Collapse
|
11
|
Effects of chloroquine and hydroxychloroquine on the sensitivity of pancreatic cancer cells to targeted therapies. Adv Biol Regul 2023; 87:100917. [PMID: 36243652 DOI: 10.1016/j.jbior.2022.100917] [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: 09/24/2022] [Accepted: 09/25/2022] [Indexed: 11/20/2022]
Abstract
Approaches to improve pancreatic cancer therapy are essential as this disease has a very bleak outcome. Approximately 80% of pancreatic cancers are pancreatic ductal adenocarcinomas (PDAC). PDAC is a cancer which is difficult to effectively treat as it is often detected late in the disease process. Almost all PDACs (over 90%) have activating mutations in the GTPase gene KRAS. These mutations result in constitutive KRas activation and the mobilization of downstream pathways such as the Raf/MEK/ERK pathway. Small molecule inhibitors of key components of the KRas/Raf/MEK/ERK pathways as well as monoclonal antibodies (MoAbs) specific for upstream growth factor receptors such insulin like growth factor-1 receptor (IGF1-R) and epidermal growth factor receptors (EGFRs) have been developed and have been evaluated in clinical trials. An additional key regulatory gene frequently mutated (∼75%) in PDAC is the TP53 tumor suppressor gene which controls the transcription of multiple genes involved in cell cycle progression, apoptosis, metabolism, cancer progression and other growth regulatory processes. Small molecule mutant TP53 reactivators have been developed which alter the structure of mutant TP53 protein and restore some of its antiproliferative activities. Some mutant TP53 reactivators have been examined in clinical trials with patients with mutant TP53 genes. Inhibitors to the TP53 negative regulator Mouse Double Minute 2 (MDM2) have been developed and analyzed in clinical trials. Chloroquine and hydroxychloroquine are established anti-malarial and anti-inflammatory drugs that also prevent the induction of autophagy which can have effects on cancer survival. Chloroquine and hydroxychloroquine have also been examined in various clinical trials. Recent studies are suggesting effective treatment of PDAC patients may require chemotherapy as well as targeting multiple pathways and biochemical processes.
Collapse
|
12
|
A Defucosylated Anti-EpCAM Monoclonal Antibody (EpMab-37-mG 2a-f) Exerts Antitumor Activity in Xenograft Model. Antibodies (Basel) 2022; 11:antib11040074. [PMID: 36546899 PMCID: PMC9774109 DOI: 10.3390/antib11040074] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/18/2022] [Accepted: 11/22/2022] [Indexed: 11/25/2022] Open
Abstract
The epithelial cell adhesion molecule (EpCAM) is a stem cell and carcinoma antigen, which mediates cellular adhesion and proliferative signaling by the proteolytic cleavage. In contrast to low expression in normal epithelium, EpCAM is frequently overexpressed in various carcinomas, which correlates with poor prognosis. Therefore, EpCAM has been considered as a promising target for tumor diagnosis and therapy. Using the Cell-Based Immunization and Screening (CBIS) method, we previously established an anti-EpCAM monoclonal antibody (EpMab-37; mouse IgG1, kappa). In this study, we investigated the antibody-dependent cellular cytotoxicity (ADCC), complement-dependent cytotoxicity (CDC), and an antitumor activity by a defucosylated mouse IgG2a-type of EpMab-37 (EpMab-37-mG2a-f) against a breast cancer cell line (BT-474) and a pancreatic cancer cell line (Capan-2), both of which express EpCAM. EpMab-37-mG2a-f recognized BT-474 and Capan-2 cells with a moderate binding-affinity [apparent dissociation constant (KD): 2.9 × 10-8 M and 1.8 × 10-8 M, respectively] by flow cytometry. EpMab-37-mG2a-f exhibited ADCC and CDC for both cells by murine splenocytes and complements, respectively. Furthermore, administration of EpMab-37-mG2a-f significantly suppressed the xenograft tumor development compared with the control mouse IgG. These results indicated that EpMab-37-mG2a-f exerts antitumor activities and could provide valuable therapeutic regimen for breast and pancreatic cancers.
Collapse
|
13
|
Liu R, He X, Li Z. Positive clinical outcomes following therapy with programmed cell death protein 1/programmed cell death ligand 1 inhibitors in neuroendocrine carcinoma of the cervix. Front Pharmacol 2022; 13:1029598. [DOI: 10.3389/fphar.2022.1029598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 11/03/2022] [Indexed: 11/18/2022] Open
Abstract
Neuroendocrine carcinoma of the cervix (NECC) is a highly aggressive and rare gynecological malignancy with a poor prognosis. Despite aggressive local and systemic treatments, there are high rates of locoregional recurrence and distant metastases. Therefore, more potent treatments are required to manage NECC. In recent years, emerging immune checkpoint inhibitors, such as programmed cell death protein 1 (PD-1)/programmed cell death ligand 1 (PD-L1) inhibitors, have been used in treating various solid tumors and provide a new direction for immune-targeted therapy for NECC. In this review, we summarize the biomarkers useful for the evaluation of the therapy with PD-1/PD-L1 inhibitors in patients with NECC and the clinical applications and prospects of monotherapy with PD-1/PD-L1 inhibitors and combinations with other therapies in patients with NECC. In some individual case reports, therapeutic strategies with PD-1/PD-L1 inhibitors showed good efficacy. Further studies are needed to confirm the possibility of using PD-1/PD-L1 inhibitors as a standard treatment strategy in NECC.
Collapse
|
14
|
Yin Z, Chen S. Therapeutic Targets and Prognostic Biomarkers Among CXC Chemokines in Pancreatic Ductal Adenocarcinoma Microenvironment. Pancreas 2022; 51:1235-1247. [PMID: 37078951 DOI: 10.1097/mpa.0000000000002178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/21/2023]
Abstract
OBJECTIVES Pancreatic ductal adenocarcinoma (PDAC) is characterized by occult onset, rapid progression, and poor prognosis. CXC chemokines play an important role in tumor microenvironment and development. However, the potential mechanistic values of CXC chemokines as clinical biomarkers and therapeutic targets in PDAC have not been fully clarified. METHODS The altered expression, interaction network, and clinical data of CXC chemokines in patients with PDAC were analyzed by the data from the Gene Expression Omnibus and the Tumor Cancer Genome Atlas. RESULTS CXCL5 transcriptional level was significantly elevated in PDAC tissues. A significant correlation was found between the expression of CXC1/3/5/8 and the pathological stage of PDAC patients. The PDAC patients with low transcriptional levels of CXCL5/9/10/11/17 were associated with a significantly better prognosis. The functions of differentially expressed CXC chemokines are primarily related to the chemokine signaling pathway, cytokine-cytokine receptor interaction, and viral protein interaction with cytokine and cytokine receptor. RELA, NFKB1, and SP1 are key transcription factors for CXC chemokines, and the SRC family of tyrosine kinases, mitogen-activated protein kinases, CDK5, PRKCQ, ROCK1, ITK, IKBKE, JAK3, and NTRK2 are CXC chemokine targets. CONCLUSIONS The results indicated that CXC chemokines might serve as therapeutic targets and prognostic biomarkers in PDAC.
Collapse
Affiliation(s)
- Zi Yin
- From the Department of General Surgery, Guangdong Provincial People's Hospital, GuangdongAcademy of Medical Sciences, Guangzhou, China
| | | |
Collapse
|
15
|
Hihara F, Matsumoto H, Yoshimoto M, Masuko T, Endo Y, Igarashi C, Tachibana T, Shinada M, Zhang MR, Kurosawa G, Sugyo A, Tsuji AB, Higashi T, Kurihara H, Ueno M, Yoshii Y. In Vitro Tumor Cell-Binding Assay to Select High-Binding Antibody and Predict Therapy Response for Personalized 64Cu-Intraperitoneal Radioimmunotherapy against Peritoneal Dissemination of Pancreatic Cancer: A Feasibility Study. Int J Mol Sci 2022; 23:5807. [PMID: 35628616 PMCID: PMC9146758 DOI: 10.3390/ijms23105807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/12/2022] [Accepted: 05/19/2022] [Indexed: 02/01/2023] Open
Abstract
Peritoneal dissemination of pancreatic cancer has a poor prognosis. We have reported that intraperitoneal radioimmunotherapy using a 64Cu-labeled antibody (64Cu-ipRIT) is a promising adjuvant therapy option to prevent this complication. To achieve personalized 64Cu-ipRIT, we developed a new in vitro tumor cell-binding assay (64Cu-TuBA) system with a panel containing nine candidate 64Cu-labeled antibodies targeting seven antigens (EGFR, HER2, HER3, TfR, EpCAM, LAT1, and CD98), which are reportedly overexpressed in patients with pancreatic cancer. We investigated the feasibility of 64Cu-TuBA to select the highest-binding antibody for individual cancer cell lines and predict the treatment response in vivo for 64Cu-ipRIT. 64Cu-TuBA was performed using six human pancreatic cancer cell lines. For three cell lines, an in vivo treatment study was performed with 64Cu-ipRIT using high-, middle-, or low-binding antibodies in each peritoneal dissemination mouse model. The high-binding antibodies significantly prolonged survival in each mouse model, while low-and middle-binding antibodies were ineffective. There was a correlation between in vitro cell binding and in vivo therapeutic efficacy. Our findings suggest that 64Cu-TuBA can be used for patient selection to enable personalized 64Cu-ipRIT. Tumor cells isolated from surgically resected tumor tissues would be suitable for analysis with the 64Cu-TuBA system in future clinical studies.
Collapse
Affiliation(s)
- Fukiko Hihara
- Department of Molecular Imaging and Theranostics, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan;
(F.H.); (H.M.); (C.I.); (T.T.); (M.S.); (M.-R.Z.); (A.S.); (A.B.T.); (T.H.)
| | - Hiroki Matsumoto
- Department of Molecular Imaging and Theranostics, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan;
(F.H.); (H.M.); (C.I.); (T.T.); (M.S.); (M.-R.Z.); (A.S.); (A.B.T.); (T.H.)
- Department of Diagnostic Radiology, Kanagawa Cancer Center, Kanagawa 241-8515, Japan;
| | - Mitsuyoshi Yoshimoto
- Division of Functional Imaging, National Cancer Center Hospital East, Chiba 277-8577, Japan;
| | - Takashi Masuko
- School of Pharmacy, Kindai University, Osaka 577-8502, Japan; (T.M.); (Y.E.)
| | - Yuichi Endo
- School of Pharmacy, Kindai University, Osaka 577-8502, Japan; (T.M.); (Y.E.)
| | - Chika Igarashi
- Department of Molecular Imaging and Theranostics, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan;
(F.H.); (H.M.); (C.I.); (T.T.); (M.S.); (M.-R.Z.); (A.S.); (A.B.T.); (T.H.)
| | - Tomoko Tachibana
- Department of Molecular Imaging and Theranostics, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan;
(F.H.); (H.M.); (C.I.); (T.T.); (M.S.); (M.-R.Z.); (A.S.); (A.B.T.); (T.H.)
| | - Mitsuhiro Shinada
- Department of Molecular Imaging and Theranostics, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan;
(F.H.); (H.M.); (C.I.); (T.T.); (M.S.); (M.-R.Z.); (A.S.); (A.B.T.); (T.H.)
- Faculty of Science, Toho University, Chiba 274-8510, Japan
| | - Ming-Rong Zhang
- Department of Molecular Imaging and Theranostics, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan;
(F.H.); (H.M.); (C.I.); (T.T.); (M.S.); (M.-R.Z.); (A.S.); (A.B.T.); (T.H.)
| | - Gene Kurosawa
- International Center for Cell and Gene Therapy, Fujita Health University, Aichi 470-1192, Japan;
| | - Aya Sugyo
- Department of Molecular Imaging and Theranostics, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan;
(F.H.); (H.M.); (C.I.); (T.T.); (M.S.); (M.-R.Z.); (A.S.); (A.B.T.); (T.H.)
| | - Atsushi B. Tsuji
- Department of Molecular Imaging and Theranostics, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan;
(F.H.); (H.M.); (C.I.); (T.T.); (M.S.); (M.-R.Z.); (A.S.); (A.B.T.); (T.H.)
| | - Tatsuya Higashi
- Department of Molecular Imaging and Theranostics, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan;
(F.H.); (H.M.); (C.I.); (T.T.); (M.S.); (M.-R.Z.); (A.S.); (A.B.T.); (T.H.)
| | - Hiroaki Kurihara
- Department of Diagnostic Radiology, Kanagawa Cancer Center, Kanagawa 241-8515, Japan;
| | - Makoto Ueno
- Department of Gastroenterology, Kanagawa Cancer Center, Kanagawa 241-8515, Japan;
| | - Yukie Yoshii
- Department of Molecular Imaging and Theranostics, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan;
(F.H.); (H.M.); (C.I.); (T.T.); (M.S.); (M.-R.Z.); (A.S.); (A.B.T.); (T.H.)
- Department of Diagnostic Radiology, Kanagawa Cancer Center, Kanagawa 241-8515, Japan;
| |
Collapse
|
16
|
Gu X, Zhou W, Han J. Factors Affecting the Readmission of Patients with Pancreatic Cancer after Surgery. Appl Bionics Biomech 2022; 2022:6106914. [PMID: 35528536 PMCID: PMC9076323 DOI: 10.1155/2022/6106914] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 04/07/2022] [Accepted: 04/18/2022] [Indexed: 12/03/2022] Open
Abstract
Objective Pancreatic cancer is one of the deadliest solid malignancies. Its surgical resection is technically very challenging and has a high risk of complications even after discharge. This study analyzed the risk factors associated with unplanned readmission after pancreatic cancer surgery. Methods Pancreatic cancer patients who were readmitted within 30 days after surgery were classified as the observation group, while those not readmitted within 30 days postsurgery were classified as the control group. The serum levels of gastrointestinal hormones, stress hormones, and peripheral immune cells of the two groups were compared at different intervals. Results No significant differences in gender and age were observed between the two groups. At 7, 14, and 21 days postsurgery, the levels of gastrointestinal hormones motilin, gastrin, calcitonin gene-related peptide, and growth hormone-releasing peptide of the observation group were lower than the control group, while the levels of adrenocorticotropin, renin, angiotensin, and plasma aldosterone of the observation group were significantly higher than the control group. In addition, compared to the control group, lower levels of CD4+T cells, CD8+T cells, and NKT cells and higher levels of Treg, Breg, and MDSC cells were observed in the peripheral blood of the observation group. Conclusion The serum levels of gastrointestinal hormones, stress hormones, and peripheral immune cells could be associated with the risk of unplanned readmission within 30 days after pancreatic cancer surgery.
Collapse
Affiliation(s)
- Xiaojing Gu
- Department of Biliary and Pancreatic Surgery, Tongji Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
- Wuhan Polytechnic University, Wuhan, Hubei 430023, China
| | - Wei Zhou
- Wuhan Polytechnic University, Wuhan, Hubei 430023, China
| | - Juan Han
- Department of Biliary and Pancreatic Surgery, Tongji Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| |
Collapse
|
17
|
Dhasmana A, Dhasmana S, Kotnala S, A A, Kashyap VK, Shaji PD, Laskar P, Khan S, Pellicano R, Fagoonee S, Haque S, Yallapu MM, Chauhan SC, Jaggi M. A topography of immunotherapies against gastrointestinal malignancies. Panminerva Med 2021; 64:56-71. [PMID: 34664484 DOI: 10.23736/s0031-0808.21.04541-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Gastrointestinal (GI) cancers are one of the leading causes of death worldwide. Although various approaches are implemented to improve the health condition of GI patients, none of the treatment protocols promise for eradicating cancer. However, a treatment mechanism against any kind of disease condition is already existing executing inside the human body. The 'immune system' is highly efficient to detect and destroy the unfavourable events of the body including tumor cells. The immune system can restrict the growth and proliferation of cancer. Cancer cells behave much smarter and adopt new mechanisms for hiding from the immune cells. Thus, cancer immunotherapy might play a decisive role to train the immune system against cancer. In this review, we have discussed the immunotherapy permitted for the treatment of GI cancers. We have discussed various methods and mechanisms, periodic development of cancer immunotherapies, approved biologicals, completed and ongoing clinical trials, role of various biopharmaceuticals, and epigenetic factors involved in GI cancer immunotherapies (graphical abstract Figure 1).
Collapse
Affiliation(s)
- Anupam Dhasmana
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX, USA.,South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA.,Department of Biosciences and Cancer Research Institute, Himalayan Institute of Medical Sciences, Swami Rama Himalayan University, Dehradun, India
| | - Swati Dhasmana
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX, USA.,South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Sudhir Kotnala
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX, USA.,South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Anukriti A
- Department of Biosciences, School of Liberal Arts and Sciences, Mody University, Lakshamgarh, Rajasthan, India
| | - Vivek K Kashyap
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX, USA.,South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Poornima D Shaji
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Partha Laskar
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX, USA.,South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Sheema Khan
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX, USA.,South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | | | - Sharmila Fagoonee
- Institute of Biostructure and Bioimaging (CNR), Molecular Biotechnology Center, Turin, Italy
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, Saudi Arabia.,Bursa Uludağ University Faculty of Medicine, Görükle Campus, Nilüfer, Bursa, Turkey
| | - Murali M Yallapu
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX, USA.,South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Subhash C Chauhan
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX, USA.,South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Meena Jaggi
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX, USA - meena.jaggi @utrgv.edu.,South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| |
Collapse
|
18
|
Schlick K, Kiem D, Greil R. Recent Advances in Pancreatic Cancer: Novel Prognostic Biomarkers and Targeted Therapy-A Review of the Literature. Biomolecules 2021; 11:1469. [PMID: 34680101 PMCID: PMC8533343 DOI: 10.3390/biom11101469] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/27/2021] [Accepted: 10/01/2021] [Indexed: 12/14/2022] Open
Abstract
Pancreatic adenocarcinoma carries a devastating prognosis. For locally advanced and metastatic disease, several chemotherapeutic regimens are currently being used. Over the past years, novel approaches have included targeting EGFR, NTRK, PARP, K-Ras as well as stroma and fibrosis, leading to approval of NTRK and PARP inhibitors. Moreover, immune check point inhibitors and different combinational approaches involving immunotherapeutic agents are being investigated in many clinical trials. MiRNAs represent a novel tool and are thought to greatly improve management by allowing for earlier diagnosis and for more precise guidance of treatment.
Collapse
Affiliation(s)
- Konstantin Schlick
- Oncologic Center, Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Paracelsus Medical University, 5020 Salzburg, Austria; (K.S.); (D.K.)
- Cancer Cluster Salzburg, 5020 Salzburg, Austria
| | - Dominik Kiem
- Oncologic Center, Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Paracelsus Medical University, 5020 Salzburg, Austria; (K.S.); (D.K.)
- Cancer Cluster Salzburg, 5020 Salzburg, Austria
| | - Richard Greil
- Oncologic Center, Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Paracelsus Medical University, 5020 Salzburg, Austria; (K.S.); (D.K.)
- Cancer Cluster Salzburg, 5020 Salzburg, Austria
- Laboratory for Immunological and Molecular Cancer Research (SCRI-LIMCR), Salzburg Cancer Research Institute, 5020 Salzburg, Austria
| |
Collapse
|