1
|
Lee D, Lee C, Han K, Goo T, Kim B, Han Y, Kwon W, Lee S, Jang JY, Park T. Machine learning models for pancreatic cancer diagnosis based on microbiome markers from serum extracellular vesicles. Sci Rep 2025; 15:10995. [PMID: 40164714 DOI: 10.1038/s41598-025-94183-y] [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: 12/16/2024] [Accepted: 03/12/2025] [Indexed: 04/02/2025] Open
Abstract
Pancreatic cancer (PC) is a fatal disease with an extremely low 5-year survival rate, mainly because of its poor detection rate in early stages. Given emerging evidence of the relationship between microbiota composition and diseases, this study aims to identify microbiome markers linked to the diagnosis of pancreatic cancer. We utilized extracellular vesicles (EVs) data obtained from blood samples of 38 pancreatic cancer patients and 51 health controls. Least absolute shrinkage and selection operator (LASSO) and stepwise method were used to obtain some candidate markers in genus and phylum levels. These markers were used to develop various machine learning models including logistic regression (LR), random forest (RF), support vector machine (SVM), and Deep Neural Network (DNN) methods. In phylum level, DNN performed best with three markers (Verrucomicrobia, Actinobacteria and Proteobacteria) selected by stepwise method with the test AUC 0.959. In genus level, DNN using 11 markers selected by LASSO (Ruminococcaceae UCG-013, Ruminiclostridium, Propionibacterium, Lachnospiraceae NK4A136 group, Corynebacterium.1, Akkermansia, Mucispirillum, Pseudomonas, Diaphorobacter, Clostridium sensu stricto 1 and Turicibacter) outperformed others with 0.961 test AUCs. These results highlight the potential of microbiome markers and prediction models in clinical studies of PC diagnosis.
Collapse
Affiliation(s)
- Doeun Lee
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, 08826, Korea
| | - Chanhee Lee
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, 08826, Korea
| | - Kyulhee Han
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, 08826, Korea
| | - Taewan Goo
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, 08826, Korea
| | - Boram Kim
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, 08826, Korea
| | - Youngmin Han
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Wooil Kwon
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Seungyeoun Lee
- Department of Applied Mathematics, Sejong University, Seoul, 03080, Korea
| | - Jin-Young Jang
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Korea.
| | - Taesung Park
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, 08826, Korea.
- Department of Statistics, Seoul National University, Seoul, 08826, Korea.
| |
Collapse
|
2
|
Breaza GM, Closca RM, Cindrea AC, Hut FE, Cretu O, Sima LV, Rakitovan M, Zara F. Immunohistochemical Evaluation of the Tumor Immune Microenvironment in Pancreatic Ductal Adenocarcinoma. Diagnostics (Basel) 2025; 15:646. [PMID: 40075893 PMCID: PMC11899021 DOI: 10.3390/diagnostics15050646] [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: 02/07/2025] [Revised: 02/27/2025] [Accepted: 03/05/2025] [Indexed: 03/14/2025] Open
Abstract
Background: Pancreatic ductal adenocarcinoma is an aggressive neoplasm with a complex carcinogenesis process that must be understood through the interactions between tumor cells and tumor microenvironment cells. Methods: This study was retrospective with a chronological extension period of 16 years and included 56 cases of pancreatic ductal adenocarcinoma. This study identified, quantified, and correlated the cells of the tumor immune microenvironment in pancreatic ductal adenocarcinoma with major prognostic factors as well as overall survival, using an extensive panel of immunohistochemical markers. Results: Three tumor immunotypes were identified: subtype A (hot immunotype), subtype B (intermediate immunotype), and subtype C (cold immunotype). Patients with immunotype C exhibit considerably higher rates of both pancreatic fistulas and acute pancreatitis. Immunotypes B and C significantly increased the risk of this complication by factors of 3.68 (p = 0.002) and 3.94 (p = 0.001), respectively. The estimated probabilities of fistula formation for each immunotype are as follows: 2.5% for immunotype A, 25% for immunotype B, and 28% for immunotype C. There was a statistically significant difference in median survival times according to tumor immunotype (p < 0.001). Specifically, patients with immunotype C tumors had a median survival time of only 120.5 days, compared to 553.5 days for those with immunotype A and 331.5 for immunotype B tumors. Conclusions: The identification of the immunotype of pancreatic ductal adenocarcinoma can be a predictive factor for the occurrence of complications such as pancreatic fistula as well as for overall survival.
Collapse
Affiliation(s)
- Gelu Mihai Breaza
- Department of Microscopic Morphology, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania; (G.M.B.); (M.R.); (F.Z.)
- University Clinic of Surgery I, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania; (F.E.H.); (O.C.); (L.V.S.)
| | - Raluca Maria Closca
- Department of Microscopic Morphology, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania; (G.M.B.); (M.R.); (F.Z.)
- Department of Pathology, Emergency City Hospital, 300254 Timisoara, Romania
| | - Alexandru Cristian Cindrea
- Department of Surgery, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania;
- Emergency Department, Emergency Clinical Municipal Hospital, 300079 Timisoara, Romania
| | - Florin Emil Hut
- University Clinic of Surgery I, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania; (F.E.H.); (O.C.); (L.V.S.)
- Center for Hepato-Bilio-Pancreatic Surgery, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania
| | - Octavian Cretu
- University Clinic of Surgery I, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania; (F.E.H.); (O.C.); (L.V.S.)
| | - Laurentiu Vasile Sima
- University Clinic of Surgery I, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania; (F.E.H.); (O.C.); (L.V.S.)
| | - Marina Rakitovan
- Department of Microscopic Morphology, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania; (G.M.B.); (M.R.); (F.Z.)
- Oro-Maxillo-Facial Surgery Clinic, Emergency City Hospital, 300062 Timisoara, Romania
| | - Flavia Zara
- Department of Microscopic Morphology, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania; (G.M.B.); (M.R.); (F.Z.)
- Department of Pathology, Emergency City Hospital, 300254 Timisoara, Romania
| |
Collapse
|
3
|
Chen X, Sun F, Wang X, Feng X, Aref AR, Tian Y, Ashrafizadeh M, Wu D. Inflammation, microbiota, and pancreatic cancer. Cancer Cell Int 2025; 25:62. [PMID: 39987122 PMCID: PMC11847367 DOI: 10.1186/s12935-025-03673-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 02/04/2025] [Indexed: 02/24/2025] Open
Abstract
Pancreatic cancer (PC) is a malignancy of gastrointestinal tract threatening the life of people around the world. In spite of the advances in the treatment of PC, the overall survival of this disease in advanced stage is less than 12%. Moreover, PC cells have aggressive behaviour in proliferation and metastasis as well as capable of developing therapy resistance. Therefore, highlighting the underlying molecular mechanisms in PC pathogenesis can provide new insights for its treatment. In the present review, inflammation and related pathways as well as role of gut microbiome in the regulation of PC pathogenesis are highlighted. The various kinds of interleukins and chemokines are able to regulate angiogenesis, metastasis, proliferation, inflammation and therapy resistance in PC cells. Furthermore, a number of molecular pathways including NF-κB, TLRs and TGF-β demonstrate dysregulation in PC aggravating inflammation and tumorigenesis. Therapeutic regulation of these pathways can reverse inflammation and progression of PC. Both chronic and acute pancreatitis have been shown to be risk factors in the development of PC, further highlighting the role of inflammation. Finally, the composition of gut microbiota can be a risk factor for PC development through affecting pathways such as NF-κB to mediate inflammation.
Collapse
Affiliation(s)
- XiaoLiang Chen
- Department of General Surgery and Integrated Traditional Chinese and Western Medicine Oncology, Tiantai People'S Hospital of Zhejiang Province(Tiantai Branch of Zhejiang Provincial People'S Hospital), Hangzhou Medical College, Taizhou, Zhejiang, China
| | - Feixia Sun
- Nursing Department, Shandong First Medical University Affiliated Occupational Disease Hospital (Shandong Provincial Occupational Disease Hospital), Jinan, China
| | - Xuqin Wang
- Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing, 401120, China
| | - Xiaoqiang Feng
- Center of Stem Cell and Regenerative Medicine, Gaozhou People's Hospital, Gaozhou, 525200, Guangdong, China
| | - Amir Reza Aref
- VitroVision Department, DeepkinetiX, Inc, Boston, MA, USA
| | - Yu Tian
- Research Center, the Huizhou Central People'S Hospital, Guangdong Medical University, Huizhou, Guangdong, China.
- School of Public Health, Benedictine University, No. 5700 College Road, Lisle, IL, 60532, USA.
| | - Milad Ashrafizadeh
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250000, Shandong, China.
| | - Dengfeng Wu
- Department of Emergency, The People'S Hospital of Gaozhou, No. 89 Xiguan Road, Gaozhou, 525200, Guangdong, China.
| |
Collapse
|
4
|
Fanijavadi S, Jensen LH. Dysbiosis-NK Cell Crosstalk in Pancreatic Cancer: Toward a Unified Biomarker Signature for Improved Clinical Outcomes. Int J Mol Sci 2025; 26:730. [PMID: 39859442 PMCID: PMC11765696 DOI: 10.3390/ijms26020730] [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: 12/18/2024] [Revised: 01/12/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive cancer with poor prognosis, primarily due to its immunosuppressive tumor microenvironment (TME), which contributes to treatment resistance. Recent research shows that the microbiome, including microbial communities in the oral cavity, gut, bile duct, and intratumoral environments, plays a key role in PDAC development, with microbial imbalances (dysbiosis) promoting inflammation, cancer progression, therapy resistance, and treatment side effects. Microbial metabolites can also affect immune cells, especially natural killer (NK) cells, which are vital for tumor surveillance, therapy response and treatment-related side effects. Dysbiosis can affect NK cell function, leading to resistance and side effects. We propose that a combined biomarker approach, integrating microbiome composition and NK cell profiles, can help predict treatment resistance and side effects, enabling more personalized therapies. This review examines how dysbiosis contributes to NK cell dysfunction in PDAC and discusses strategies (e.g., antibiotics, probiotics, vaccines) to modulate the microbiome and enhance NK cell function. Targeting dysbiosis could modulate NK cell activity, improve the effectiveness of PDAC treatments, and reduce side effects. However, further research is needed to develop unified NK cell-microbiome interaction-based biomarkers for more precise and effective patient outcomes.
Collapse
Affiliation(s)
- Sara Fanijavadi
- Cancer Polyclinic, Levanger Hospital, 7601 Levanger, Trøndelag, Norway
- Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark;
| | - Lars Henrik Jensen
- Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark;
- Department of Oncology, Institute of Regional Health Research, University of Southern Denmark, 7100 Vejle, Denmark
| |
Collapse
|
5
|
Uguz A, Muftuoglu C, Mert U, Gumus T, Ece D, Asadi M, Bagci OU, Caner A. Unveiling Microbiota Profiles in Saliva and Pancreatic Tissues of Patients with Pancreatic Cancer. Microorganisms 2025; 13:119. [PMID: 39858887 PMCID: PMC11767796 DOI: 10.3390/microorganisms13010119] [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: 11/23/2024] [Revised: 12/08/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025] Open
Abstract
The pancreas, previously considered a sterile organ, has recently been shown to harbor its own microbiota that may influence tumor biology and patient outcomes. Despite increasing interest in the impact of the microbiome on cancer, the relationship between pancreatic tissue and oral microbiomes in pancreatic ductal adenocarcinoma (PDAC) remains limited. In this study, the oral and pancreas tissue microbiomes of patients with PDAC were compared to patients with other periampullary cancers (DC/AC) and a healthy control group using 16S rRNA gene sequence analysis. The results showed a significant reduction in microbial diversity in the saliva of cancer patients compared to healthy controls, while the PDAC patients exhibited a distinct microbial profile in their pancreatic tissues, consisting predominantly of Firmicutes, Proteobacteria, and Actinobacter, after filtering the microbiome of the indoor environment. Notably, the presence of oral bacteria such as Anoxybacillus, Clostridium, and Bacillus in pancreatic tissues suggests potential translocation from the oral cavity. This study emphasizes the importance of understanding the role of body fluid and tissue microbiota in pancreatic cancer, proposing that oral dysbiosis may contribute to disease progression. Moreover, the results suggest that the microbiome of the indoor environment in which samples are collected and analyzed is also important in microbiota analysis studies.
Collapse
Affiliation(s)
- Alper Uguz
- Department of General Surgery, Faculty of Medicine, Ege University, 35040 Izmir, Turkey; (A.U.); (T.G.)
| | - Can Muftuoglu
- Department of Basic Oncology, Institute of Health Sciences, Ege University, 35100 Izmir, Turkey; (C.M.); (D.E.); (M.A.); (O.U.B.)
- Department of Medicine I, University Medical Centre Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Ufuk Mert
- Atatürk Vocational School of Health Services, Ege University, 35100 Izmir, Turkey;
- Translational Pulmonary Research Center (EGESAM), Ege University, 35100 Izmir, Turkey
| | - Tufan Gumus
- Department of General Surgery, Faculty of Medicine, Ege University, 35040 Izmir, Turkey; (A.U.); (T.G.)
| | - Deniz Ece
- Department of Basic Oncology, Institute of Health Sciences, Ege University, 35100 Izmir, Turkey; (C.M.); (D.E.); (M.A.); (O.U.B.)
| | - Milad Asadi
- Department of Basic Oncology, Institute of Health Sciences, Ege University, 35100 Izmir, Turkey; (C.M.); (D.E.); (M.A.); (O.U.B.)
| | - Ozlem Ulusan Bagci
- Department of Basic Oncology, Institute of Health Sciences, Ege University, 35100 Izmir, Turkey; (C.M.); (D.E.); (M.A.); (O.U.B.)
- Department of Microbiology, Faculty of Medicine, Ankara University, 06230 Ankara, Turkey
| | - Ayse Caner
- Department of Basic Oncology, Institute of Health Sciences, Ege University, 35100 Izmir, Turkey; (C.M.); (D.E.); (M.A.); (O.U.B.)
- Translational Pulmonary Research Center (EGESAM), Ege University, 35100 Izmir, Turkey
- Department of Parasitology, Faculty of Medicine, Ege University, 35100 Izmir, Turkey
| |
Collapse
|
6
|
Şeulean EC, Dumitraşcu DL. The association between exocrine pancreatic insufficiency and changes in gut microbiota: a narrative review. Med Pharm Rep 2025; 98:5-12. [PMID: 39949904 PMCID: PMC11817577 DOI: 10.15386/mpr-2638] [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: 05/04/2023] [Revised: 07/25/2023] [Accepted: 08/07/2023] [Indexed: 02/16/2025] Open
Abstract
Due to their physical proximity, the healthy pancreas and the gut microbiome are known to interact in a variety of ways. The gut microbiota has been recognized as a potential factor in the development and progression of exocrine pancreatic insufficiency through several mechanisms. Pancreatic diseases like chronic and acute pancreatitis or pancreatic cancer are frequently accompanied by pancreatic exocrine insufficiency which affects the gut microbiota. Firstly, the gut microbes are controlled by antimicrobial pancreatic secretions, while themselves induce the secretion of substances by the pancreas through metabolite production, such as short-chain fatty acids. Secondly, dysbiosis, the alteration in the abundance and diversity of different species, has been observed in patients with pancreatic diseases. Dysbiosis influences carcinogenesis in pancreatic cancer in ways that are either procarcinogenic or anticarcinogenic and finding these connections will have clinical implications. Identifying microbial biomarkers allow for an earlier diagnosis, improved therapy and prognosis in pancreatic cancer. The gut microbiome has a role in the pathogenesis of pancreatitis by either a bacterial translocation or a host immune response mechanism. The disruption of the normal gut barrier is believed to be the primary source of bacteria in acute pancreatitis which leads to infected pancreatic necrosis. In this paper, we review the current data about the association between pancreatic diseases linked to exocrine insufficiency and gut microbiota.
Collapse
Affiliation(s)
- Edina C Şeulean
- 2 Department of Internal Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Dan L Dumitraşcu
- 2 Department of Internal Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| |
Collapse
|
7
|
Arif T, Nazir F, Aurangzeb RF, Hussain M, Aurangzeb RI, Rehman A, Kumar K, Islam R, Islam H, Khalid Q, Arrey Agbor DB, Munir K, Bokhari SFH, Shehryar A, Ibrahim M. The Potential of Fecal and Urinary Biomarkers for Early Detection of Pancreatic Ductal Adenocarcinoma: A Systematic Review. Cureus 2024; 16:e59248. [PMID: 38813271 PMCID: PMC11134185 DOI: 10.7759/cureus.59248] [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] [Accepted: 04/28/2024] [Indexed: 05/31/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal cancer often diagnosed at advanced stages, highlighting the urgent need for early detection strategies. This systematic review explores the potential of fecal and urinary biomarkers for early PDAC detection. A comprehensive search identified eight relevant studies investigating various biomarkers, including proteins, metabolites, microbial profiles, DNA mutations, and non-coding RNAs. Promising findings suggest that urinary biomarkers related to metabolic alterations, inflammatory processes, fecal microbiome profiles, and fecal miRNAs hold diagnostic potential even at early stages of PDAC. Combining biomarkers into panels may enhance diagnostic accuracy. Challenges such as validation in larger cohorts, standardization of protocols, and regulatory approval must be addressed for clinical translation. Despite these hurdles, non-invasive urinary and fecal biomarkers represent a promising avenue for improving PDAC outcomes through early detection.
Collapse
Affiliation(s)
- Talha Arif
- Accident and Emergency, Imran Idrees Teaching Hospital, Sialkot, PAK
| | - Faran Nazir
- Internal Medicine, Faisalabad Medical University, Deer Park, USA
| | | | | | | | | | - Kabeer Kumar
- Internal Medicine, Chandka Medical College, Larkana, PAK
| | - Rabia Islam
- Research, Faisalabad Medical University, Faisalabad, PAK
| | - Hamza Islam
- Internal Medicine, Punjab Medical College, Faisalabad, PAK
| | - Qais Khalid
- Internal Medicine, Khyber Medical University, Peshawar, PAK
| | | | - Kashaf Munir
- Medicine, Shalamar Medical and Dental College, Lahore, PAK
| | | | | | | |
Collapse
|
8
|
Lupu VV, Bratu RM, Trandafir LM, Bozomitu L, Paduraru G, Gimiga N, Ghiga G, Forna L, Ioniuc I, Petrariu FD, Puha B, Lupu A. Exploring the Microbial Landscape: Gut Dysbiosis and Therapeutic Strategies in Pancreatitis-A Narrative Review. Biomedicines 2024; 12:645. [PMID: 38540258 PMCID: PMC10967871 DOI: 10.3390/biomedicines12030645] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/09/2024] [Accepted: 03/12/2024] [Indexed: 01/03/2025] Open
Abstract
The gut microbiota is emerging as an important contributor to the homeostasis of the human body through its involvement in nutrition and metabolism, protection against pathogens, and the development and modulation of the immune system. It has therefore become an important research topic in recent decades. Although the association between intestinal dysbiosis and numerous digestive pathologies has been thoroughly researched, its involvement in pancreatic diseases constitutes a novelty in the specialized literature. In recent years, growing evidence has pointed to the critical involvement of the pancreas in regulating the intestinal microbiota, as well as the impact of the intestinal microbiota on pancreatic physiology, which implies the existence of a bidirectional connection known as the "gut-pancreas axis". It is theorized that any change at either of these levels triggers a response in the other component, hence leading to the evolution of pancreatitis. However, there are not enough data to determine whether gut dysbiosis is an underlying cause or a result of pancreatitis; therefore, more research is needed in this area. The purpose of this narrative review is to highlight the role of gut dysbiosis in the pathogenesis of acute and chronic pancreatitis, its evolution, and the prospect of employing the microbiota as a therapeutic intervention for pancreatitis.
Collapse
Affiliation(s)
| | - Roxana Mihaela Bratu
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (V.V.L.); (L.M.T.); (L.B.); (N.G.); (G.G.); (L.F.); (I.I.); (F.D.P.); (B.P.); (A.L.)
| | | | | | - Gabriela Paduraru
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (V.V.L.); (L.M.T.); (L.B.); (N.G.); (G.G.); (L.F.); (I.I.); (F.D.P.); (B.P.); (A.L.)
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Szczepanski JM, Rudolf MA, Shi J. Clinical Evaluation of the Pancreatic Cancer Microenvironment: Opportunities and Challenges. Cancers (Basel) 2024; 16:794. [PMID: 38398185 PMCID: PMC10887250 DOI: 10.3390/cancers16040794] [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: 01/16/2024] [Revised: 02/10/2024] [Accepted: 02/12/2024] [Indexed: 02/25/2024] Open
Abstract
Advances in our understanding of pancreatic ductal adenocarcinoma (PDAC) and its tumor microenvironment (TME) have the potential to transform treatment for the hundreds of thousands of patients who are diagnosed each year. Whereas the clinical assessment of cancer cell genetics has grown increasingly sophisticated and personalized, current protocols to evaluate the TME have lagged, despite evidence that the TME can be heterogeneous within and between patients. Here, we outline current protocols for PDAC diagnosis and management, review novel biomarkers, and highlight potential opportunities and challenges when evaluating the PDAC TME as we prepare to translate emerging TME-directed therapies to the clinic.
Collapse
Affiliation(s)
| | | | - Jiaqi Shi
- Department of Pathology and Clinical Labs, University of Michigan, Ann Arbor, MI 48109, USA; (J.M.S.); (M.A.R.)
| |
Collapse
|
10
|
Horvat NK, Karpovsky I, Phillips M, Wyatt MM, Hall MA, Herting CJ, Hammons J, Mahdi Z, Moffitt RA, Paulos CM, Lesinski GB. Clinically relevant orthotopic pancreatic cancer models for adoptive T cell transfer therapy. J Immunother Cancer 2024; 12:e008086. [PMID: 38191243 PMCID: PMC10806555 DOI: 10.1136/jitc-2023-008086] [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] [Accepted: 12/18/2023] [Indexed: 01/10/2024] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is an aggressive tumor. Prognosis is poor and survival is low in patients diagnosed with this disease, with a survival rate of ~12% at 5 years. Immunotherapy, including adoptive T cell transfer therapy, has not impacted the outcomes in patients with PDAC, due in part to the hostile tumor microenvironment (TME) which limits T cell trafficking and persistence. We posit that murine models serve as useful tools to study the fate of T cell therapy. Currently, genetically engineered mouse models (GEMMs) for PDAC are considered a "gold-standard" as they recapitulate many aspects of human disease. However, these models have limitations, including marked tumor variability across individual mice and the cost of colony maintenance. METHODS Using flow cytometry and immunohistochemistry, we characterized the immunological features and trafficking patterns of adoptively transferred T cells in orthotopic PDAC (C57BL/6) models using two mouse cell lines, KPC-Luc and MT-5, isolated from C57BL/6 KPC-GEMM (KrasLSL-G12D/+p53-/- and KrasLSL-G12D/+p53LSL-R172H/+, respectively). RESULTS The MT-5 orthotopic model best recapitulates the cellular and stromal features of the TME in the PDAC GEMM. In contrast, far more host immune cells infiltrate the KPC-Luc tumors, which have less stroma, although CD4+ and CD8+ T cells were similarly detected in the MT-5 tumors compared with KPC-GEMM in mice. Interestingly, we found that chimeric antigen receptor (CAR) T cells redirected to recognize mesothelin on these tumors that signal via CD3ζ and 41BB (Meso-41BBζ-CAR T cells) infiltrated the tumors of mice bearing stroma-devoid KPC-Luc orthotopic tumors, but not MT-5 tumors. CONCLUSIONS Our data establish for the first time a reproducible and realistic clinical system useful for modeling stroma-rich and stroma-devoid PDAC tumors. These models shall serve an indepth study of how to overcome barriers that limit antitumor activity of adoptively transferred T cells.
Collapse
Affiliation(s)
- Natalie K Horvat
- Department of Pediatric Hematology, Oncology and Immunology, Emory University, Atlanta, Georgia, USA
| | - Isaac Karpovsky
- Department of Hematology and Oncology, Emory University, Atlanta, Georgia, USA
| | - Maggie Phillips
- Department of Hematology and Oncology, Emory University, Atlanta, Georgia, USA
| | - Megan M Wyatt
- Department of Surgery, Department of Microbiology & Immunology, Emory University Winship Cancer Institute, Atlanta, Georgia, USA
| | - Margaret A Hall
- Department of Hematology and Oncology, Emory University, Atlanta, Georgia, USA
| | - Cameron J Herting
- Department of Hematology and Oncology, Emory University, Atlanta, Georgia, USA
| | - Jacklyn Hammons
- Department of Hematology and Oncology, Emory University, Atlanta, Georgia, USA
| | - Zaid Mahdi
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA
| | - Richard A Moffitt
- Department of Hematology and Oncology, Emory University, Atlanta, Georgia, USA
| | - Chrystal M Paulos
- Department of Surgery, Department of Microbiology & Immunology, Emory University Winship Cancer Institute, Atlanta, Georgia, USA
| | - Gregory B Lesinski
- Department of Hematology and Oncology, Emory University Winship Cancer Institute, Atlanta, Georgia, USA
| |
Collapse
|
11
|
Merali N, Chouari T, Terroire J, Jessel MD, Liu DSK, Smith JH, Wooldridge T, Dhillon T, Jiménez JI, Krell J, Roberts KJ, Rockall TA, Velliou E, Sivakumar S, Giovannetti E, Demirkan A, Annels NE, Frampton AE. Bile Microbiome Signatures Associated with Pancreatic Ductal Adenocarcinoma Compared to Benign Disease: A UK Pilot Study. Int J Mol Sci 2023; 24:16888. [PMID: 38069211 PMCID: PMC10706407 DOI: 10.3390/ijms242316888] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/18/2023] [Accepted: 11/24/2023] [Indexed: 12/18/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a very poor survival. The intra-tumoural microbiome can influence pancreatic tumourigenesis and chemoresistance and, therefore, patient survival. The role played by bile microbiota in PDAC is unknown. We aimed to define bile microbiome signatures that can effectively distinguish malignant from benign tumours in patients presenting with obstructive jaundice caused by benign and malignant pancreaticobiliary disease. Prospective bile samples were obtained from 31 patients who underwent either Endoscopic Retrograde Cholangiopancreatography (ERCP) or Percutaneous Transhepatic Cholangiogram (PTC). Variable regions (V3-V4) of the 16S rRNA genes of microorganisms present in the samples were amplified by Polymerase Chain Reaction (PCR) and sequenced. The cohort consisted of 12 PDAC, 10 choledocholithiasis, seven gallstone pancreatitis and two primary sclerosing cholangitis patients. Using the 16S rRNA method, we identified a total of 135 genera from 29 individuals (12 PDAC and 17 benign). The bile microbial beta diversity significantly differed between patients with PDAC vs. benign disease (Permanova p = 0.0173). The separation of PDAC from benign samples is clearly seen through unsupervised clustering of Aitchison distance. We found three genera to be of significantly lower abundance among PDAC samples vs. benign, adjusting for false discovery rate (FDR). These were Escherichia (FDR = 0.002) and two unclassified genera, one from Proteobacteria (FDR = 0.002) and one from Enterobacteriaceae (FDR = 0.011). In the same samples, the genus Streptococcus (FDR = 0.033) was found to be of increased abundance in the PDAC group. We show that patients with obstructive jaundice caused by PDAC have an altered microbiome composition in the bile compared to those with benign disease. These bile-based microbes could be developed into potential diagnostic and prognostic biomarkers for PDAC and warrant further investigation.
Collapse
Affiliation(s)
- Nabeel Merali
- Minimal Access Therapy Training Unit (MATTU), Royal Surrey County Hospital NHS Foundation Trust, Egerton Road, Guildford GU2 7XX, UK
- Department of Hepato-Pancreato-Biliary (HPB) Surgery, Royal Surrey County Hospital NHS Foundation Trust, Egerton Road, Guildford GU2 7XX, UK
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford GU2 7WG, UK
| | - Tarak Chouari
- Department of Hepato-Pancreato-Biliary (HPB) Surgery, Royal Surrey County Hospital NHS Foundation Trust, Egerton Road, Guildford GU2 7XX, UK
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford GU2 7WG, UK
| | - Julien Terroire
- Surrey Institute for People-Centred AI, University of Surrey, Guildford GU2 7XH, UK
- Section of Statistical Multi-Omics, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford GU2 7WG, UK
| | - Maria-Danae Jessel
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford GU2 7WG, UK
| | - Daniel S. K. Liu
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, UK
| | - James-Halle Smith
- Hepatobiliary and Pancreatic Surgery Unit, Queen Elizabeth Hospital Birmingham, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TH, UK
| | - Tyler Wooldridge
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford GU2 7WG, UK
| | - Tony Dhillon
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford GU2 7WG, UK
| | - José I. Jiménez
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Jonathan Krell
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, UK
| | - Keith J. Roberts
- Hepatobiliary and Pancreatic Surgery Unit, Queen Elizabeth Hospital Birmingham, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TH, UK
| | - Timothy A. Rockall
- Minimal Access Therapy Training Unit (MATTU), Royal Surrey County Hospital NHS Foundation Trust, Egerton Road, Guildford GU2 7XX, UK
| | - Eirini Velliou
- Centre for 3D Models of Health and Disease, Division of Surgery and Interventional Science, University College London (UCL), London W1W 7TY, UK
| | - Shivan Sivakumar
- Oncology Department, Institute of Immunology and Immunotherapy, Birmingham Medical School, University of Birmingham, Birmingham B15 2TT, UK
| | - Elisa Giovannetti
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
- Cancer Pharmacology Lab, AIRC Start Up Unit, Fondazione Pisana per la Scienza, San Giuliano Terme PI, 56017 Pisa, Italy
| | - Ayse Demirkan
- Surrey Institute for People-Centred AI, University of Surrey, Guildford GU2 7XH, UK
- Section of Statistical Multi-Omics, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford GU2 7WG, UK
| | - Nicola E. Annels
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford GU2 7WG, UK
| | - Adam E. Frampton
- Minimal Access Therapy Training Unit (MATTU), Royal Surrey County Hospital NHS Foundation Trust, Egerton Road, Guildford GU2 7XX, UK
- Department of Hepato-Pancreato-Biliary (HPB) Surgery, Royal Surrey County Hospital NHS Foundation Trust, Egerton Road, Guildford GU2 7XX, UK
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford GU2 7WG, UK
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, UK
| |
Collapse
|
12
|
Oldfield L, Costello E. Where the metabolome meets the microbiome for pancreatic cancer detection. Cell Rep Med 2023; 4:101011. [PMID: 37729875 PMCID: PMC10518497 DOI: 10.1016/j.xcrm.2023.101011] [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: 09/22/2023]
Abstract
Risk prediction tools for pancreatic cancer are urgently sought to facilitate screening. Irajizad et al.1 describe the performance of a risk predication model based on circulating microbial- and non-microbial metabolites for assessment of 5-year pancreatic cancer risk.
Collapse
Affiliation(s)
- Lucy Oldfield
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Eithne Costello
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK.
| |
Collapse
|
13
|
Henderson EA, Lukomski S, Boone BA. Emerging applications of cancer bacteriotherapy towards treatment of pancreatic cancer. Front Oncol 2023; 13:1217095. [PMID: 37588093 PMCID: PMC10425600 DOI: 10.3389/fonc.2023.1217095] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 06/26/2023] [Indexed: 08/18/2023] Open
Abstract
Pancreatic cancer is a highly aggressive form of cancer with a five-year survival rate of only ten percent. Pancreatic ductal adenocarcinoma (PDAC) accounts for ninety percent of those cases. PDAC is associated with a dense stroma that confers resistance to current treatment modalities. Increasing resistance to cancer treatments poses a challenge and a need for alternative therapies. Bacterial mediated cancer therapies were proposed in the late 1800s by Dr. William Coley when he injected osteosarcoma patients with live streptococci or a fabrication of heat-killed Streptococcus pyogenes and Serratia marcescens known as Coley's toxin. Since then, several bacteria have gained recognition for possible roles in potentiating treatment response, enhancing anti-tumor immunity, and alleviating adverse effects to standard treatment options. This review highlights key bacterial mechanisms and structures that promote anti-tumor immunity, challenges and risks associated with bacterial mediated cancer therapies, and applications and opportunities for use in PDAC management.
Collapse
Affiliation(s)
- Emily A. Henderson
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, WV, United States
| | - Slawomir Lukomski
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, WV, United States
- West Virginia Cancer Institute, West Virginia University, Morgantown, WV, United States
| | - Brian A. Boone
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, WV, United States
- West Virginia Cancer Institute, West Virginia University, Morgantown, WV, United States
- Department of Surgery, West Virginia University, Morgantown, WV, United States
| |
Collapse
|
14
|
Kumar L, Kumar S, Sandeep K, Patel SKS. Therapeutic Approaches in Pancreatic Cancer: Recent Updates. Biomedicines 2023; 11:1611. [PMID: 37371705 DOI: 10.3390/biomedicines11061611] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 05/25/2023] [Accepted: 05/27/2023] [Indexed: 06/29/2023] Open
Abstract
Cancer is a significant challenge for effective treatment due to its complex mechanism, different progressing stages, and lack of adequate procedures for screening and identification. Pancreatic cancer is typically identified in its advanced progression phase with a low survival of ~5 years. Among cancers, pancreatic cancer is also considered a high mortality-causing casualty over other accidental or disease-based mortality, and it is ranked seventh among all mortality-associated cancers globally. Henceforth, developing diagnostic procedures for its early detection, understanding pancreatic cancer-linked mechanisms, and various therapeutic strategies are crucial. This review describes the recent development in pancreatic cancer progression, mechanisms, and therapeutic approaches, including molecular techniques and biomedicines for effectively treating cancer.
Collapse
Affiliation(s)
- Lokender Kumar
- School of Biotechnology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan 173229, India
| | - Sanjay Kumar
- Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Greater Noida 201310, India
| | - Kumar Sandeep
- Dr. B.R.A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi 110029, India
| | | |
Collapse
|
15
|
Ijichi H. Significance of Tumor Microenvironment for Regulating Pancreatic Cancer. Cancers (Basel) 2023; 15:cancers15092482. [PMID: 37173948 PMCID: PMC10177064 DOI: 10.3390/cancers15092482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
Pancreatic cancer is the most lethal common cancer in the world [...].
Collapse
Affiliation(s)
- Hideaki Ijichi
- Clinical Nutrition Center, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| |
Collapse
|
16
|
Bhoopathi P, Mannangatti P, Das SK, Fisher PB, Emdad L. Chemoresistance in pancreatic ductal adenocarcinoma: Overcoming resistance to therapy. Adv Cancer Res 2023; 159:285-341. [PMID: 37268399 DOI: 10.1016/bs.acr.2023.02.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC), a prominent cause of cancer deaths worldwide, is a highly aggressive cancer most frequently detected at an advanced stage that limits treatment options to systemic chemotherapy, which has provided only marginal positive clinical outcomes. More than 90% of patients with PDAC die within a year of being diagnosed. PDAC is increasing at a rate of 0.5-1.0% per year, and it is expected to be the second leading cause of cancer-related mortality by 2030. The resistance of tumor cells to chemotherapeutic drugs, which can be innate or acquired, is the primary factor contributing to the ineffectiveness of cancer treatments. Although many PDAC patients initially responds to standard of care (SOC) drugs they soon develop resistance caused partly by the substantial cellular heterogeneity seen in PDAC tissue and the tumor microenvironment (TME), which are considered key factors contributing to resistance to therapy. A deeper understanding of molecular mechanisms involved in PDAC progression and metastasis development, and the interplay of the TME in all these processes is essential to better comprehend the etiology and pathobiology of chemoresistance observed in PDAC. Recent research has recognized new therapeutic targets ushering in the development of innovative combinatorial therapies as well as enhancing our comprehension of several different cell death pathways. These approaches facilitate the lowering of the therapeutic threshold; however, the possibility of subsequent resistance development still remains a key issue and concern. Discoveries, that can target PDAC resistance, either alone or in combination, have the potential to serve as the foundation for future treatments that are effective without posing undue health risks. In this chapter, we discuss potential causes of PDAC chemoresistance and approaches for combating chemoresistance by targeting different pathways and different cellular functions associated with and mediating resistance.
Collapse
Affiliation(s)
- Praveen Bhoopathi
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Richmond, VA, United States
| | - Padmanabhan Mannangatti
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Richmond, VA, United States
| | - Swadesh K Das
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.
| | - Luni Emdad
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.
| |
Collapse
|
17
|
Fudalej M, Kwaśniewska D, Nurzyński P, Badowska-Kozakiewicz A, Mękal D, Czerw A, Sygit K, Deptała A. New Treatment Options in Metastatic Pancreatic Cancer. Cancers (Basel) 2023; 15:cancers15082327. [PMID: 37190255 DOI: 10.3390/cancers15082327] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/12/2023] [Accepted: 04/15/2023] [Indexed: 05/17/2023] Open
Abstract
Pancreatic cancer (PC) is the seventh leading cause of cancer death across the world. Poor prognosis of PC is associated with several factors, such as diagnosis at an advanced stage, early distant metastases, and remarkable resistance to most conventional treatment options. The pathogenesis of PC seems to be significantly more complicated than originally assumed, and findings in other solid tumours cannot be extrapolated to this malignancy. To develop effective treatment schemes prolonging patient survival, a multidirectional approach encompassing different aspects of the cancer is needed. Particular directions have been established; however, further studies bringing them all together and connecting the strengths of each therapy are needed. This review summarises the current literature and provides an overview of new or emerging therapeutic strategies for the more effective management of metastatic PC.
Collapse
Affiliation(s)
- Marta Fudalej
- Department of Oncology Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland
- Department of Oncology, Central Clinical Hospital of the Ministry of Interior and Administration, 02-507 Warsaw, Poland
| | - Daria Kwaśniewska
- Department of Oncology, Central Clinical Hospital of the Ministry of Interior and Administration, 02-507 Warsaw, Poland
| | - Paweł Nurzyński
- Department of Oncology, Central Clinical Hospital of the Ministry of Interior and Administration, 02-507 Warsaw, Poland
| | | | - Dominika Mękal
- Department of Oncology Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland
| | - Aleksandra Czerw
- Department of Health Economics and Medical Law, Medical University of Warsaw, 01-445 Warsaw, Poland
- Department of Economic and System Analyses, National Institute of Public Health NIH-National Research Institute, 00-791 Warsaw, Poland
| | - Katarzyna Sygit
- Faculty of Health Sciences, Calisia University, 62-800 Kalisz, Poland
| | - Andrzej Deptała
- Department of Oncology Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland
- Department of Oncology, Central Clinical Hospital of the Ministry of Interior and Administration, 02-507 Warsaw, Poland
| |
Collapse
|