1
|
Hassan MS, Johnson C, Ponna S, Scofield D, Awasthi N, von Holzen U. Inhibition of Insulin-like Growth Factor 1 Receptor/Insulin Receptor Signaling by Small-Molecule Inhibitor BMS-754807 Leads to Improved Survival in Experimental Esophageal Adenocarcinoma. Cancers (Basel) 2024; 16:3175. [PMID: 39335147 PMCID: PMC11430189 DOI: 10.3390/cancers16183175] [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: 08/12/2024] [Revised: 09/04/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
The insulin-like growth factor-1 (IGF-1) and insulin axes are upregulated in obesity and obesity-associated esophageal adenocarcinoma (EAC). Nanoparticle albumin-bound paclitaxel (nab-paclitaxel) is a contemporary nanotechnology-based paclitaxel (PT) bound to human albumin, ensuring its solubility in water rather than a toxic solvent. Here, we examined the benefits of inhibiting insulin-like growth factor-1 receptor/insulin receptor (IGF-1/IR) signaling and the enhancement of nab-paclitaxel effects by inclusion of the small-molecule inhibitor BMS-754807 using both in vitro and in vivo models of EAC. Using multiple EAC cell lines, BMS-754807 and nab-paclitaxel were evaluated as mono and combination therapies for in vitro effects on cell proliferation, cell death, and cell movement. We then analyzed the in vivo anticancer potency with survival improvement with BMS-754807 and nab-paclitaxel mono and combination therapies. BMS-754807 monotherapy suppressed in vitro cell proliferation and wound healing while increasing apoptosis. BMS-754807, when combined with nab-paclitaxel, enhanced those effects on the inhibition of cell proliferation, increment in cell apoptosis, and inhibition of wound healing. BMS-754807 with nab-paclitaxel produced substantially greater antitumor effects by increasing in vivo apoptosis, leading to increased mice survival compared to those of BMS-754807 or nab-paclitaxel monotherapy. Our outcomes support the use of BMS-754807, alone and in combination with nab-paclitaxel, as an efficient and innovative treatment choice for EAC.
Collapse
Affiliation(s)
- Md Sazzad Hassan
- Department of Surgery, Indiana University School of Medicine, South Bend, IN 46617, USA; (N.A.)
- Harper Cancer Research Institute, South Bend, IN 46617, USA
| | - Chloe Johnson
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Saisantosh Ponna
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Dimitri Scofield
- Department of Biology, Indiana University, South Bend, IN 47405, USA
| | - Niranjan Awasthi
- Department of Surgery, Indiana University School of Medicine, South Bend, IN 46617, USA; (N.A.)
- Harper Cancer Research Institute, South Bend, IN 46617, USA
| | - Urs von Holzen
- Department of Surgery, Indiana University School of Medicine, South Bend, IN 46617, USA; (N.A.)
- Harper Cancer Research Institute, South Bend, IN 46617, USA
- Goshen Center for Cancer Care, Goshen, IN 46526, USA
- School of Medicine, University of Basel, 4056 Basel, Switzerland
| |
Collapse
|
2
|
Hassan MS, Cwidak N, Johnson C, Däster S, Eppenberger-Castori S, Awasthi N, Li J, Schwarz MA, von Holzen U. Therapeutic Potential of the Cyclin-Dependent Kinase Inhibitor Flavopiridol on c-Myc Overexpressing Esophageal Cancer. Front Pharmacol 2021; 12:746385. [PMID: 34621175 PMCID: PMC8490822 DOI: 10.3389/fphar.2021.746385] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 09/08/2021] [Indexed: 12/14/2022] Open
Abstract
Tumors with elevated c-Myc expression often exhibit a highly aggressive phenotype, and c-Myc amplification has been shown to be frequent in esophageal cancer. Emerging data suggests that synthetic lethal interactions between c-Myc pathway activation and small molecules inhibition involved in cell cycle signaling can be therapeutically exploited to preferentially kill tumor cells. We therefore investigated whether exploiting elevated c-Myc expression is effective in treating esophageal cancer with the CDK inhibitor flavopiridol. We found frequent overexpression of c-Myc in human esophageal cancer cell lines and tissues. c-Myc overexpression correlated with accelerated esophageal cancer subcutaneous xenograft tumor growth. Esophageal cancer cells with elevated c-Myc expression were found preferentially more sensitive to induction of apoptosis by the CDK inhibition flavopiridol compared to esophageal cancer cells with lower c-Myc expression. In addition, we observed that flavopiridol alone or in combination with the chemotherapeutic agent nanoparticle albumin-bound paclitaxel (NPT) or in combinations with the targeted agent BMS-754807 significantly inhibited esophageal cancer cell proliferation and subcutaneous xenograft tumor growth while significantly enhancing overall mice survival. These results indicate that aggressive esophageal cancer cells with elevated c-Myc expression are sensitive to the CDK inhibitor flavopiridol, and that flavopiridol alone or in combination can be a potential therapy for c-Myc overexpressing esophageal cancer.
Collapse
Affiliation(s)
- Md Sazzad Hassan
- Department of Surgery, Indiana University School of Medicine, South Bend, IN, United States.,Harper Cancer Research Institute, South Bend, IN, United States
| | - Nicholas Cwidak
- Department of Surgery, Indiana University School of Medicine, South Bend, IN, United States
| | - Chloe Johnson
- University of Notre Dame, Notre Dame, IN, United States
| | | | | | - Niranjan Awasthi
- Department of Surgery, Indiana University School of Medicine, South Bend, IN, United States.,Harper Cancer Research Institute, South Bend, IN, United States
| | - Jun Li
- Harper Cancer Research Institute, South Bend, IN, United States.,University of Notre Dame, Notre Dame, IN, United States
| | - Margaret A Schwarz
- Harper Cancer Research Institute, South Bend, IN, United States.,Department of Pediatrics, Indiana University School of Medicine, South Bend, IN, United States
| | - Urs von Holzen
- Department of Surgery, Indiana University School of Medicine, South Bend, IN, United States.,Harper Cancer Research Institute, South Bend, IN, United States.,University of Basel, Basel, Switzerland.,Goshen Center for Cancer Care, Goshen, IN, United States
| |
Collapse
|
3
|
Shademan B, Karamad V, Nourazarian A, Avcı CB. CAR T Cells: Cancer Cell Surface Receptors Are the Target for Cancer Therapy. Adv Pharm Bull 2021; 12:476-489. [PMID: 35935042 PMCID: PMC9348524 DOI: 10.34172/apb.2022.051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 06/12/2021] [Accepted: 08/17/2021] [Indexed: 11/09/2022] Open
Abstract
Immunotherapy has become a prominent strategy for the treatment of cancer. A method that improves the immune system's ability to attack a tumor (Enhances antigen binding). Targeted killing of malignant cells by adoptive transfer of chimeric antigen receptor (CAR) T cells is a promising immunotherapy technique in the treatment of cancers. For this purpose, the patient's immune cells, with genetic engineering aid, are loaded with chimeric receptors that have particular antigen binding and activate cytotoxic T lymphocytes. That increases the effectiveness of immune cells and destroying cancer cells. This review discusses the basic structure and function of CAR-T cells and how antigenic targets are identified to treat different cancers and address the disadvantages of this treatment for cancer.
Collapse
Affiliation(s)
- Behrouz Shademan
- Department of Medical Biology, Faculty of Medicine, EGE University, Izmir, Turkey
| | - Vahidreza Karamad
- Department of Medical Biology, Faculty of Medicine, EGE University, Izmir, Turkey
| | - Alireza Nourazarian
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Cigir Biray Avcı
- Department of Medical Biology, Faculty of Medicine, EGE University, Izmir, Turkey
| |
Collapse
|
4
|
Fuentes-Baile M, Ventero MP, Encinar JA, García-Morales P, Poveda-Deltell M, Pérez-Valenciano E, Barberá VM, Gallego-Plazas J, Rodríguez-Lescure Á, Martín-Nieto J, Saceda M. Differential Effects of IGF-1R Small Molecule Tyrosine Kinase Inhibitors BMS-754807 and OSI-906 on Human Cancer Cell Lines. Cancers (Basel) 2020; 12:cancers12123717. [PMID: 33322337 PMCID: PMC7763458 DOI: 10.3390/cancers12123717] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 12/09/2020] [Indexed: 01/30/2023] Open
Abstract
Simple Summary We have tested the effects of IGF-1R tyrosine kinase inhibitors BMS-754807 (BMS) and OSI-906 (OSI) on human colon, pancreatic carcinoma cell, and glioblastoma cell lines and primary cultures. Although OSI and BMS are able to inhibit IGF-1R activity at low doses, the differential effect on cell proliferation and cell-cycle phase distribution shown by both compounds probes that many effects observed are mediated by BMS off-target interactions. Using MAPKs ELISAs and phospho-RTK array analysis, we have identified several BMS regulated putative kinases able to mediate BMS off-target effects. Interestingly, molecular docking assays suggest that BMS could affect these kinases not only by blocking their ATP-binding domain, but also by means of allosteric interactions. Since BMS has an important antineoplastic effect on these poor prognosis types of cancer, these compounds could be taken in consideration for treatment independently of IGF-1R status. Abstract We have determined the effects of the IGF-1R tyrosine kinase inhibitors BMS-754807 (BMS) and OSI-906 (OSI) on cell proliferation and cell-cycle phase distribution in human colon, pancreatic carcinoma, and glioblastoma cell lines and primary cultures. IGF-1R signaling was blocked by BMS and OSI at equivalent doses, although both inhibitors exhibited differential antiproliferative effects. In all pancreatic carcinoma cell lines tested, BMS exerted a strong antiproliferative effect, whereas OSI had a minimal effect. Similar results were obtained on glioblastoma primary cultures, where HGUE-GB-15, -16 and -17 displayed resistance to OSI effects, whereas they were inhibited in their proliferation by BMS. Differential effects of BMS and OSI were also observed in colon carcinoma cell lines. Both inhibitors also showed different effects on cell cycle phase distribution, BMS induced G2/M arrest followed by cell death, while OSI induced G1 arrest with no cell death. Both inhibitors also showed different effects on other protein kinases activities. Taken together, our results are indicative that BMS mainly acts through off-target effects exerted on other protein kinases. Given that BMS exhibits a potent antiproliferative effect, we believe that this compound could be useful for the treatment of different types of tumors independently of their IGF-1R activation status.
Collapse
Affiliation(s)
- María Fuentes-Baile
- Unidad de Investigación, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO), Hospital General Universitario de Elche, 03203 Elche (Alicante), Spain; (M.F.-B.); (V.M.B.)
| | - María P. Ventero
- Unidad de Investigación, Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Hospital General Universitario de Alicante, 03005 Alicante, Spain;
| | - José A. Encinar
- Instituto de Biología Molecular y Celular (IBMC) and Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, 03202 Elche (Alicante), Spain; (P.G.-M.); (M.P.-D.); (E.P.-V.)
- Correspondence: (J.A.E.); (M.S.); Tel.: +34-966658432 (M.S.)
| | - Pilar García-Morales
- Instituto de Biología Molecular y Celular (IBMC) and Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, 03202 Elche (Alicante), Spain; (P.G.-M.); (M.P.-D.); (E.P.-V.)
| | - María Poveda-Deltell
- Instituto de Biología Molecular y Celular (IBMC) and Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, 03202 Elche (Alicante), Spain; (P.G.-M.); (M.P.-D.); (E.P.-V.)
| | - Elizabeth Pérez-Valenciano
- Instituto de Biología Molecular y Celular (IBMC) and Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, 03202 Elche (Alicante), Spain; (P.G.-M.); (M.P.-D.); (E.P.-V.)
| | - Víctor M. Barberá
- Unidad de Investigación, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO), Hospital General Universitario de Elche, 03203 Elche (Alicante), Spain; (M.F.-B.); (V.M.B.)
- Unidad de Genética Molecular, Hospital General Universitario de Elche, 03203 Elche (Alicante), Spain
| | - Javier Gallego-Plazas
- Servicio de Oncología, Hospital General Universitario de Elche, 03203 Elche (Alicante), Spain; (J.G.-P.); (Á.R.-L.)
| | - Álvaro Rodríguez-Lescure
- Servicio de Oncología, Hospital General Universitario de Elche, 03203 Elche (Alicante), Spain; (J.G.-P.); (Á.R.-L.)
| | - José Martín-Nieto
- Departamento de Fisiología, Genética y Microbiología, Facultad de Ciencias, Universidad de Alicante, 03080 Alicante, Spain;
| | - Miguel Saceda
- Unidad de Investigación, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO), Hospital General Universitario de Elche, 03203 Elche (Alicante), Spain; (M.F.-B.); (V.M.B.)
- Instituto de Biología Molecular y Celular (IBMC) and Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, 03202 Elche (Alicante), Spain; (P.G.-M.); (M.P.-D.); (E.P.-V.)
- Correspondence: (J.A.E.); (M.S.); Tel.: +34-966658432 (M.S.)
| |
Collapse
|
5
|
Elaileh A, Saharia A, Potter L, Baio F, Ghafel A, Abdelrahim M, Heyne K. Promising new treatments for pancreatic cancer in the era of targeted and immune therapies. Am J Cancer Res 2019; 9:1871-1888. [PMID: 31598392 PMCID: PMC6780661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 08/05/2019] [Indexed: 06/10/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer mortality among men and women in the United States. Its incidence has been on the rise, with a projected two-fold increase by 2030. PDAC carries a poor prognosis due to a lack of effective screening tools, limited understanding of pathophysiology, and ineffective treatment modalities. Recently, there has been a revolution in the world of oncology with the advent of novel treatments to combat this disease. However, the 5-year survival of PDAC remains unchanged at a dismal 8%. The aim of this review is to bring together several studies and identify various recent modalities that have been promising in treating PDAC.
Collapse
Affiliation(s)
- Ahmed Elaileh
- Department of General Surgery, Houston Methodist HospitalHouston, Texas, USA
| | - Ashish Saharia
- Department of General Surgery, Houston Methodist HospitalHouston, Texas, USA
| | - Lucy Potter
- Department of General Surgery, Houston Methodist HospitalHouston, Texas, USA
| | - Flavio Baio
- Department of General Surgery, Houston Methodist HospitalHouston, Texas, USA
| | - Afnan Ghafel
- Department of Radiology, The University of JordanAmman, Jordan
| | - Maen Abdelrahim
- Department of General Surgery, Houston Methodist HospitalHouston, Texas, USA
| | - Kirk Heyne
- Department of General Surgery, Houston Methodist HospitalHouston, Texas, USA
| |
Collapse
|
6
|
Wang Z, Li Y, Zhou F, Piao Z, Hao J. β-elemene enhances anticancer bone neoplasms efficacy of paclitaxel through regulation of GPR124 in bone neoplasms cells. Oncol Lett 2018; 16:2143-2150. [PMID: 30008912 PMCID: PMC6036473 DOI: 10.3892/ol.2018.8909] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Accepted: 05/24/2018] [Indexed: 12/19/2022] Open
Abstract
The purpose of the present study was to investigate the anticancer effects of β-elemene and paclitaxel for bone neoplasms. MTT assay, reverse transcription-quantitative polymerase chain reaction, western blotting, flow cytometry and immunostaining were used to analyze the combined effects of β-elemene and paclitaxel both in vitro and in vivo. The results demonstrated that combined treatment of β-elemene and paclitaxel (β-elemene-paclitaxel) significantly inhibited growth and aggressiveness of U-2OS cells compared with either β-elemene or paclitaxel treatment alone. It was demonstrated that β-elemene promoted paclitaxel-induced apoptosis of U-2OS cells. Anti-apoptosis B-cell lymphoma (Bcl)-2 and Bcl-w genes were downregulated and pro-apoptotic Bcl-2-associated agonist of cell death and caspase-3 genes were upregulated in U-2OS cells following treatment with β-elemene-paclitaxel. Treatment of β-elemene-paclitaxel arrested the cell cycle and decreased cyclin-dependent kinase, cyclin-B1, P21 and P27 expression levels and decreased resistant genes alterations of ATP binding cassette subfamily B member 1, LDL receptor related protein and TS in U-2OS cells. Results demonstrated that β-elemene-paclitaxel decreased G-protein coupled receptor 124 (GPR124), vascular endothelial growth factor receptor, matrix metallopeptidase (MMP)-3, MMP-9 expression levels and increased endostatin, TIMP metallopeptidase inhibitor (TIMP)-1, TIMP-2 expression in U-2OS cells. In vivo assay demonstrated that β-elemene-paclitaxel treatment inhibited tumor growth of BALB/c-nu/nu nude mice and prolonged survival rate of tumor-bearing mice. Immunostaining demonstrated that β-elemene-paclitaxel treatment increased apoptotic bodies, GPR124 and increased endostatin, TIMP-1 and TIMP-2 expression in tumor tissues. In conclusion, these results suggest that the combined treatment of β-elemene-paclitaxel is more effective at inhibiting bone neoplasm growth than β-elemene or paclitaxel single treatment GPR124.
Collapse
Affiliation(s)
- Zongze Wang
- Department of Orthopedics, Nankai Hospital of Tianjin, Tianjin 300100, P.R. China
| | - Ying Li
- Renal Department of Internal Medicine, The Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Fengxin Zhou
- Department of Orthopedics, Nankai Hospital of Tianjin, Tianjin 300100, P.R. China
| | - Zhe Piao
- Department of Orthopedics, Nankai Hospital of Tianjin, Tianjin 300100, P.R. China
| | - Jian Hao
- Department of Orthopedics, Nankai Hospital of Tianjin, Tianjin 300100, P.R. China
| |
Collapse
|
7
|
Pseudomonas aeruginosa-mannose-sensitive hemagglutinin inhibits pancreatic cancer cell proliferation and induces apoptosis via the EGFR pathway and caspase signaling. Oncotarget 2018; 7:77916-77925. [PMID: 27788491 PMCID: PMC5363631 DOI: 10.18632/oncotarget.12844] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 10/14/2016] [Indexed: 12/16/2022] Open
Abstract
Pseudomonas aeruginosa-mannose-sensitive hemagglutinin (PA-MSHA) has demonstrated efficacy against several solid tumors. In this study, we found that PA-MSHA inhibited the proliferation of PANC-1 and SW1990 pancreatic cancer cells, but had no obvious effects on HPDE6-C7 normal human pancreatic duct epithelial cells. Electron microscopy revealed the presence of apoptotic bodies and intracellular vacuole formation in PA-MSHA-treated pancreatic cancer cells. Flow cytometric analysis indicated the rate of apoptosis correlated with the PA-MSHA concentration. We observed a decrease in cell fractions in G0/G1 and G2/M phases, and an increase in the fraction in S phase (p < 0.01). PA-MSHA thus caused cell cycle arrest. Increasing concentrations of PA-MSHA did not alter total levels of EGFR, AKT or ERK, but levels of the corresponding phosphoproteins decreased. PA-MSHA also reduced tumor volume in a xenograft mouse model of pancreatic cancer (p < 0.01). Furthermore, caspase-3 levels decreased while the levels of cleaved caspase-3 increased (p < 0.01). These data suggest that by blocking cell cycle progression, PA-MSHA induces apoptosis and inhibits tumor growth. PA-MSHA-mediated inhibition of EGFR signaling and activation of the caspase pathway may play an important role in the induction of apoptosis in pancreatic cancer cells.
Collapse
|
8
|
Superior Therapeutic Efficacy of Nanoparticle Albumin Bound Paclitaxel Over Cremophor-Bound Paclitaxel in Experimental Esophageal Adenocarcinoma. Transl Oncol 2018; 11:426-435. [PMID: 29475139 PMCID: PMC5884213 DOI: 10.1016/j.tranon.2018.01.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 01/18/2018] [Accepted: 01/18/2018] [Indexed: 12/18/2022] Open
Abstract
Esophageal adenocarcinoma (EAC) is the fastest growing cancer in the western world and the overall 5 year survival rate of EAC is below 20%. Most patients with EAC present with locally advanced or widespread metastatic disease, where current treatment is largely ineffective. Therefore, new therapeutic approaches are urgently needed. Nanoparticle albumin-bound paclitaxel (nab-paclitaxel) is a novel albumin-stabilized, cremophor-free and water soluble nanoparticle formulation of paclitaxel, and the potential role of nab-paclitaxel has not been tested yet in experimental EAC. Here we tested the antiproliferative and antitumor efficacy with survival advantage of nab-paclitaxel as monotherapy and in combinations in in-vitro, and in murine subcutaneous xenograft and peritoneal metastatic survival models of human EAC. Nab-paclitaxel significantly inhibited in-vitro cell proliferation with higher in-vivo antitumour efficacy and survival benefit compared to paclitaxel or carboplatin treatments both in mono- and combination therapies. Nab-paclitaxel treatment increased expression of mitotic-spindle associated phospho-stathmin, decreased expression of proliferative markers and enhanced apoptosis. This study demonstrates that nab-paclitaxel had stronger antiproliferative and antitumor activity in experimental EAC than the current standard chemotherapeutic agents which supports the rationale for its clinical use in EAC.
Collapse
|
9
|
Giordano G, Pancione M, Olivieri N, Parcesepe P, Velocci M, Di Raimo T, Coppola L, Toffoli G, D’Andrea MR. Nano albumin bound-paclitaxel in pancreatic cancer: Current evidences and future directions. World J Gastroenterol 2017; 23:5875-5886. [PMID: 28932079 PMCID: PMC5583572 DOI: 10.3748/wjg.v23.i32.5875] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 07/03/2017] [Accepted: 08/01/2017] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer (PDAC) is an aggressive and chemoresistant disease, representing the fourth cause of cancer related deaths in western countries. Majority of patients have unresectable, locally advanced or metastatic disease at time of diagnosis and the 5-year survival rate in these conditions is extremely low. For more than a decade gemcitabine has been the cornerstone of metastatic PDAC treatment, although survival benefit was very poor. PDAC cells are surrounded by an intense desmoplastic reaction that may create a barrier to the drugs penetration within the tumor. Recently PDAC stroma has been addressed as a potential therapeutic target. Nano albumin bound (Nab)-paclitaxel is an innovative molecule depleting tumor stroma, through interaction between albumin and secreted protein acidic and rich in cysteine. Addition of nab-paclitaxel to gemcitabine has showed activity and efficacy in metastatic PDAC first-line treatment improving survival and overall response rate vs gemcitabine alone in the MPACT phase III study. This combination represents one of the standards of care in advanced PDAC therapy and is suitable to a broader spectrum of patients compared to other schedules. Nab-paclitaxel is under investigation as a backbone of chemotherapy in novel combinations with target agents or immunotherapy in locally advanced or metastatic PDAC. In this article, we provide an updated and critical overview about the role of nab-paclitaxel in PDAC treatment based on the latest advances in preclinical and clinical research. Furthermore, we focus on the use of nab-paclitaxel within the context of metastatic PDAC treatment landscape and we discuss about future implications in the light of current clinical ongoing trials.
Collapse
Affiliation(s)
- Guido Giordano
- Medical Oncology Unit, San Filippo Neri Hospital, 00135 Roma, Italy
- CRO Aviano National Cancer Institute, 33081 Aviano, Italy
| | - Massimo Pancione
- Department of Sciences and Technologies, University of Sannio, 82100 Benevento, Italy
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, Complutense University, 28040 Madrid, Spain
| | - Nunzio Olivieri
- Department of Biology, University of Naples, Federico II, Via Mezzocannone, 80134 Napoli, Italy
| | - Pietro Parcesepe
- Department of Pathology and Diagnostics, University of Verona Strada, 37134 Verona, Italy
| | - Marianna Velocci
- Medical Oncology Unit, San Filippo Neri Hospital, 00135 Roma, Italy
| | - Tania Di Raimo
- Medical Oncology Unit, San Filippo Neri Hospital, 00135 Roma, Italy
| | - Luigi Coppola
- Anatomic Pathology Unit, San Filippo Neri, 00135 Roma, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, CRO-National Cancer Institute Via F, 33081 Aviano (Pordenone), Italy
| | | |
Collapse
|
10
|
Zeleniak AE, Huang W, Brinkman MK, Fishel ML, Hill R. Loss of MTSS1 results in increased metastatic potential in pancreatic cancer. Oncotarget 2017; 8:16473-16487. [PMID: 28146435 PMCID: PMC5369978 DOI: 10.18632/oncotarget.14869] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Accepted: 01/19/2017] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a 5-year survival rate of 7%. This dismal prognosis is largely due to the inability to diagnose the disease before metastasis occurs. Tumor cell dissemination occurs early in PDAC. While it is known that inflammation facilitates this process, the underlying mechanisms responsible for this progression have not been fully characterized. Here, we functionally test the role of metastasis suppressor 1 (MTSS1) in PDAC. Despite evidence showing that MTSS1 could be important for regulating metastasis in many different cancers, its function in PDAC has not been studied. Here, we show that loss of MTSS1 leads to increased invasion and migration in PDAC cell lines. Moreover, PDAC cells treated with cancer-associated fibroblast-conditioned media also have increased metastatic potential, which is augmented by loss of MTSS1. Finally, overexpression of MTSS1 in PDAC cell lines leads to a loss of migratory potential in vitro and an increase in overall survival in vivo. Collectively, our data provide insight into an important role for MTSS1 in suppressing tumor cell invasion and migration driven by the tumor microenvironment and suggest that therapeutic strategies aimed at increasing MTSS1 levels may effectively slow the development of metastatic lesions, increasing survival of patients with PDAC.
Collapse
Affiliation(s)
- Ann E Zeleniak
- Integrated Biomedical Sciences Program, University of Notre Dame, South Bend, Indiana, USA.,Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana, USA
| | - Wei Huang
- Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana, USA.,Department of Biological Sciences, University of Notre Dame, South Bend, Indiana, USA
| | - Mary K Brinkman
- Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana, USA.,Department of Biological Sciences, University of Notre Dame, South Bend, Indiana, USA
| | - Melissa L Fishel
- Indiana University School of Medicine, Department of Pharmacology and Toxicology, Indianapolis, Indiana, USA.,Indiana University School of Medicine, Department of Pediatrics, Wells Center for Pediatric Research, Indianapolis, Indiana, USA.,Pancreatic Cancer Signature Center, Indianapolis, Indiana, USA
| | - Reginald Hill
- Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana, USA.,Department of Biological Sciences, University of Notre Dame, South Bend, Indiana, USA
| |
Collapse
|
11
|
A novel intraperitoneal metastatic xenograft mouse model for survival outcome assessment of esophageal adenocarcinoma. PLoS One 2017; 12:e0171824. [PMID: 28225784 PMCID: PMC5321464 DOI: 10.1371/journal.pone.0171824] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 01/26/2017] [Indexed: 01/15/2023] Open
Abstract
Esophageal adenocarcinoma (EAC) has become the dominant type of esophageal cancer in United States. The 5-year survival rate of EAC is below 20% and most patients present with locally advanced or widespread metastatic disease, where current treatment is largely ineffective. Therefore, new therapeutic approaches are urgently needed. Improvement of EAC patient outcome requires well-characterized animal models in which to evaluate novel therapeutics. In this study we aimed to establish a peritoneal dissemination xenograft mouse model of EAC that would support survival outcome analyses. To find the best candidate cell line from 7 human EAC cell lines of different origin named ESO26, OE33, ESO51, SK-GT-2, OE19, OACM5.1C and Flo-1 were injected intraperitoneally/subcutaneously into SCID mice. The peritoneal/xenograft tumor formation and mouse survival were compared among different groups. All cell lines injected subcutaneously formed tumors within 3 months at variable rates. All cell lines except OACM5.1C formed intraperitoneal tumors within 3 months at variable rates. Median animal survival with peritoneal dissemination was 108 days for ESO26 cells (5X106), 65 days for OE33 cells (5X106), 88 days for ESO51 cells (5X106), 76 days for SK-GT-2 cells (5X106), 55 days for OE19 cells (5X106), 45 days for OE19 cells (10X106) and 82 days for Flo-1 cells (5X106). Interestingly, only in the OE19 model all mice (7/7 for 5X106 and 5/5 for10X106) developed bloody ascites with liver metastasis after intraperitoneal injection. The median survival time of these animals was the shortest (45 days for 10X106 cells). In addition, median survival was significantly increased after paclitaxel treatment compared with the control group (57 days versus 45 days, p = 0.0034) along with a significant decrease of the relative subcutaneous tumor volume (p = 0.00011). Thus peritoneal dissemination mouse xenograft model for survival outcome assessment after intraperitoneal injection of OE19 cells will be very useful for the evaluation of cancer therapeutics.
Collapse
|