1
|
Eldehna WM, Tawfik HO, Abdulla MH, Nafie MS, Aref H, Shaldam MA, Alhassan NS, Al Obeed O, Elsayed ZM, Abdel-Aziz HA. Identification of indole-grafted pyrazolopyrimidine and pyrazolopyridine derivatives as new anti-cancer agents: Synthesis, biological assessments, and molecular modeling insights. Bioorg Chem 2024; 153:107804. [PMID: 39276491 DOI: 10.1016/j.bioorg.2024.107804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/26/2024] [Accepted: 09/03/2024] [Indexed: 09/17/2024]
Abstract
In the current medical era, developing new PIM-1 inhibitors stands as a significant approach to cancer management due to the pivotal role of PIM-1 kinase in promoting cell survival, proliferation, and drug resistance in various cancers. This study involved designing and synthesizing new derivatives of pyrazolo[1,5-a]pyrimidines (6a-i) and pyrazolo[3,4-b]pyridines (10a-i) as potential anti-cancer agents targeting PIM-1 kinase. The cytotoxicity was screened on three cancer cell lines: A-549 (lung), PANC-1 (pancreatic), and A-431 (skin), alongside MRC5 normal lung cells to assess selectivity. Several pyrazolo[1,5-a]pyrimidines (6b, 6c, 6g, 6h, and 6i) and pyrazolo[3,4-b]pyridine (10f) demonstrated notable anticancer properties, particularly against A-549 lung cancer cells (IC50 range: 1.28-3.52 μM), also they exhibited significantly lower toxicity towards MRC5 normal cells. Thereafter, the compounds were evaluated for their inhibitory activity against PIM-1 kinase. Notably, 10f, bearing a 4-methoxyphenyl moiety, demonstrated good inhibition of PIM-1 with an IC50 of 0.18 μM. Additionally, 10f induced apoptosis and arrested cell cycle progression in A-549 cells. Molecular docking and dynamics simulations provided insights into the binding interactions and compounds' stability with PIM-1 kinase. The results highlight these compounds, especially 10f, as promising selective anticancer agents targeting PIM-1 kinase.
Collapse
Affiliation(s)
- Wagdy M Eldehna
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, P.O. Box 33516, Egypt; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Pharos University in Alexandria, Canal El Mahmoudia St., Alexandria 21648, Egypt.
| | - Haytham O Tawfik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt.
| | - Maha-Hamadien Abdulla
- Colorectal Research Chair, Department of Surgery, College of Medicine, King Saud University, Riyadh 11472, Saudi Arabia.
| | - Mohamed S Nafie
- Department of Chemistry, College of Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; Chemistry Department, Faculty of Science, Suez Canal University, Ismailia 41522, Egypt.
| | - Heba Aref
- Medicinal Chemistry Department, Faculty of Pharmacy, El Menoufia University, El Menoufia, Shebin El Kom 32511, Egypt
| | - Moataz A Shaldam
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, P.O. Box 33516, Egypt
| | - Noura S Alhassan
- Colorectal Research Chair, Department of Surgery, College of Medicine, King Saud University, Riyadh 11472, Saudi Arabia
| | - Omar Al Obeed
- Colorectal Research Chair, Department of Surgery, College of Medicine, King Saud University, Riyadh 11472, Saudi Arabia
| | - Zainab M Elsayed
- Scientific Research and Innovation Support Unit, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Hatem A Abdel-Aziz
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Pharos University in Alexandria, Canal El Mahmoudia St., Alexandria 21648, Egypt; Department of Applied Organic Chemistry, National Research Center, Dokki, Cairo 12622, Egypt
| |
Collapse
|
2
|
Choudhury R, Bahadi CK, Ray IP, Dash P, Pattanaik I, Mishra S, Mohapatra SR, Patnaik S, Nikhil K. PIM1 kinase and its diverse substrate in solid tumors. Cell Commun Signal 2024; 22:529. [PMID: 39487435 PMCID: PMC11531143 DOI: 10.1186/s12964-024-01898-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 10/18/2024] [Indexed: 11/04/2024] Open
Abstract
The PIM kinase family, consisting of PIM1, PIM2, and PIM3, is a group of serine/threonine protein kinases crucial for cellular growth, immunoregulation, and oncogenesis. PIM1 kinase is often overexpressed in solid and hematopoietic malignancies, promoting cell survival, proliferation, migration, and senescence by activating key genes. In vitro and in vivo studies have established the oncogenic potential of PIM1 kinases. These kinases have been implicated in tumor progression, metastasis, and resistance to chemotherapy, underscoring their potential as a therapeutic target for cancer therapy. This review delves into the intricate molecular mechanisms through which PIM1 interacts with specific substrates in different tumor tissues, leading to diverse outcomes in various human cancers. Over the past decade, the inhibition of PIM1 in cancers has garnered significant attention as a potential standalone treatment. Various in vitro, in vivo, and early clinical trial data have provided support for this approach to varying extents. Novel compounds that inhibit PIM1 kinase have shown effectiveness and a favorable toxicity profile in preclinical studies. Several of these substances are now being studied in clinical trials due to their promising outcomes. This article provides a thorough examination of the PIM1 kinase pathways and the recent advancements in producing PIM1 kinase inhibitors for the treatment of cancer.
Collapse
Affiliation(s)
- Rituparna Choudhury
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed-to-Be-University, Bhubaneswar, 751024, India
| | - Chandan Kumar Bahadi
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed-to-Be-University, Bhubaneswar, 751024, India
| | - Ipsa Pratibimbita Ray
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed-to-Be-University, Bhubaneswar, 751024, India
| | - Pragyanshree Dash
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed-to-Be-University, Bhubaneswar, 751024, India
| | - Isha Pattanaik
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed-to-Be-University, Bhubaneswar, 751024, India
| | - Suman Mishra
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed-to-Be-University, Bhubaneswar, 751024, India
| | - Soumya R Mohapatra
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed-to-Be-University, Bhubaneswar, 751024, India
| | - Srinivas Patnaik
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed-to-Be-University, Bhubaneswar, 751024, India
| | - Kumar Nikhil
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed-to-Be-University, Bhubaneswar, 751024, India.
| |
Collapse
|
3
|
Torres-Ayuso P, Katerji M, Mehlich D, Lookingbill SA, Sabbasani VR, Liou H, Casillas AL, Chauhan SS, Serwa R, Rubin MR, Marusiak AA, Swenson RE, Warfel NA, Brognard J. PIM1 targeted degradation prevents the emergence of chemoresistance in prostate cancer. Cell Chem Biol 2024; 31:326-337.e11. [PMID: 38016478 PMCID: PMC10922308 DOI: 10.1016/j.chembiol.2023.10.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 08/08/2023] [Accepted: 10/29/2023] [Indexed: 11/30/2023]
Abstract
PIM kinases have important pro-tumorigenic roles and mediate several oncogenic traits, including cell proliferation, survival, and chemotherapeutic resistance. As a result, multiple PIM inhibitors have been pursued as investigational new drugs in cancer; however, response to PIM inhibitors in solid tumors has fallen short of expectations. We found that inhibition of PIM kinase activity stabilizes protein levels of all three PIM isoforms (PIM1/2/3), and this can promote resistance to PIM inhibitors and chemotherapy. To overcome this effect, we designed PIM proteolysis targeting chimeras (PROTACs) to target PIM for degradation. PIM PROTACs effectively downmodulated PIM levels through the ubiquitin-proteasome pathway. Importantly, degradation of PIM kinases was more potent than inhibition of catalytic activity at inducing apoptosis in prostate cancer cell line models. In conclusion, we provide evidence of the advantages of degrading PIM kinases versus inhibiting their catalytic activity to target the oncogenic functions of PIM kinases.
Collapse
Affiliation(s)
- Pedro Torres-Ayuso
- Laboratory of Cell and Developmental Signaling, National Cancer Institute, NIH, Frederick, MD 21702, USA; Department of Cancer and Cellular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Meghri Katerji
- Laboratory of Cell and Developmental Signaling, National Cancer Institute, NIH, Frederick, MD 21702, USA
| | - Dawid Mehlich
- Laboratory of Cell and Developmental Signaling, National Cancer Institute, NIH, Frederick, MD 21702, USA; Laboratory of Molecular OncoSignalling, IMol Polish Academy of Sciences, 00-783 Warsaw, Poland; Doctoral School of the Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Sophia A Lookingbill
- Laboratory of Cell and Developmental Signaling, National Cancer Institute, NIH, Frederick, MD 21702, USA
| | - Venkata R Sabbasani
- Chemistry and Synthesis Center, National Heart Lung and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Hope Liou
- University of Arizona Cancer Center, Tucson, AZ 85724, USA; Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724, USA
| | | | - Shailender S Chauhan
- University of Arizona Cancer Center, Tucson, AZ 85724, USA; Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Remigiusz Serwa
- ReMedy International Research Agenda Unit, IMol Polish Academy of Sciences, 00-783 Warsaw, Poland
| | - Maxine R Rubin
- Laboratory of Cell and Developmental Signaling, National Cancer Institute, NIH, Frederick, MD 21702, USA
| | - Anna A Marusiak
- Laboratory of Molecular OncoSignalling, IMol Polish Academy of Sciences, 00-783 Warsaw, Poland
| | - Rolf E Swenson
- Chemistry and Synthesis Center, National Heart Lung and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Noel A Warfel
- University of Arizona Cancer Center, Tucson, AZ 85724, USA; Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724, USA.
| | - John Brognard
- Laboratory of Cell and Developmental Signaling, National Cancer Institute, NIH, Frederick, MD 21702, USA.
| |
Collapse
|
4
|
Chen S, Yang Y, Yuan Y, Bo Liu. Targeting PIM kinases in cancer therapy: An update on pharmacological small-molecule inhibitors. Eur J Med Chem 2024; 264:116016. [PMID: 38071792 DOI: 10.1016/j.ejmech.2023.116016] [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: 04/27/2023] [Revised: 07/15/2023] [Accepted: 11/28/2023] [Indexed: 12/30/2023]
Abstract
PIM kinases, a serine/threonine kinase family with three isoforms, has been well-known to participate in multiple physiological processes by phosphorylating various downstream targets. Accumulating evidence has recently unveiled that aberrant upregulation of PIM kinases (PIM1, PIM2, and PIM3) are closely associated with tumor cell proliferation, migration, survival, and even resistance. Inhibiting or silencing of PIM kinases has been reported have remarkable antitumor effects, such as anti-proliferation, pro-apoptosis and resensitivity, indicating the therapeutic potential of PIM kinases as potential druggable targets in many types of human cancers. More recently, several pharmacological small-molecule inhibitors have been preclinically and clinically evaluated and showed their therapeutic potential; however, none of them has been approved for clinical application so far. Thus, in this perspective, we focus on summarizing the oncogenic roles of PIM kinases, key signaling network, and pharmacological small-molecule inhibitors, which will provide a new clue on discovering more candidate antitumor drugs targeting PIM kinases in the future.
Collapse
Affiliation(s)
- Siwei Chen
- Department of Thoracic Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yushang Yang
- Department of Thoracic Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yong Yuan
- Department of Thoracic Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Bo Liu
- Department of Thoracic Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
5
|
Stosic K, Senar OA, Tarfouss J, Bouchart C, Navez J, Van Laethem JL, Arsenijevic T. A Comprehensive Review of the Potential Role of Liquid Biopsy as a Diagnostic, Prognostic, and Predictive Biomarker in Pancreatic Ductal Adenocarcinoma. Cells 2023; 13:3. [PMID: 38201207 PMCID: PMC10778087 DOI: 10.3390/cells13010003] [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: 10/30/2023] [Revised: 12/13/2023] [Accepted: 12/15/2023] [Indexed: 01/12/2024] Open
Abstract
Pancreatic ductal adenocarcinoma is one of the most lethal malignant diseases, with a mortality rate being close to incidence. Due to its heterogeneity and plasticity, as well as the lack of distinct symptoms in the early phases, it is very often diagnosed at an advanced stage, resulting in poor prognosis. Traditional tissue biopsies remain the gold standard for making a diagnosis, but have an obvious disadvantage in their inapplicability for frequent sampling. Blood-based biopsies represent a non-invasive method which potentially offers easy and repeated sampling, leading to the early detection and real-time monitoring of the disease and hopefully an accurate prognosis. Given the urgent need for a reliable biomarker that can estimate a patient's condition and response to an assigned treatment, blood-based biopsies are emerging as a potential new tool for improving patients' survival and surveillance. In this article, we discuss the current advances and challenges in using liquid biopsies for pancreatic cancer, focusing on circulating tumour DNA (ctDNA), extracellular vesicles (EVs), and circulating tumour cells (CTCs), and compare the performance and reliability of different biomarkers and combinations of biomarkers.
Collapse
Affiliation(s)
- Kosta Stosic
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, 1070 Brussels, Belgium (O.A.S.); (C.B.)
| | - Oier Azurmendi Senar
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, 1070 Brussels, Belgium (O.A.S.); (C.B.)
| | - Jawad Tarfouss
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, 1070 Brussels, Belgium (O.A.S.); (C.B.)
| | - Christelle Bouchart
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, 1070 Brussels, Belgium (O.A.S.); (C.B.)
- Department of Radiation Oncology, Hopital Universitaire de Bruxelles (H.U.B.), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Julie Navez
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, 1070 Brussels, Belgium (O.A.S.); (C.B.)
- Department of Hepato-Biliary-Pancreatic Surgery, Hopital Universitaire de Bruxelles (H.U.B.), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Jean-Luc Van Laethem
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, 1070 Brussels, Belgium (O.A.S.); (C.B.)
- Department of Gastroenterology, Hepatology and Digestive Oncology, Hopital Universitaire de Bruxelles (H.U.B.), Université Libre de Bruxelles (ULB), Route de Lennik 808, 1070 Brussels, Belgium
| | - Tatjana Arsenijevic
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, 1070 Brussels, Belgium (O.A.S.); (C.B.)
- Department of Gastroenterology, Hepatology and Digestive Oncology, Hopital Universitaire de Bruxelles (H.U.B.), Université Libre de Bruxelles (ULB), Route de Lennik 808, 1070 Brussels, Belgium
| |
Collapse
|
6
|
Ingle K, LaComb JF, Graves LM, Baines AT, Bialkowska AB. AUM302, a novel triple kinase PIM/PI3K/mTOR inhibitor, is a potent in vitro pancreatic cancer growth inhibitor. PLoS One 2023; 18:e0294065. [PMID: 37943821 PMCID: PMC10635512 DOI: 10.1371/journal.pone.0294065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 10/24/2023] [Indexed: 11/12/2023] Open
Abstract
Pancreatic cancer is one of the leading causes of cancer deaths, with pancreatic ductal adenocarcinoma (PDAC) being the most common subtype. Advanced stage diagnosis of PDAC is common, causing limited treatment opportunities. Gemcitabine is a frequently used chemotherapeutic agent which can be used as a monotherapy or in combination. However, tumors often develop resistance to gemcitabine. Previous studies show that the proto-oncogene PIM kinases (PIM1 and PIM3) are upregulated in PDAC compared to matched normal tissue and are related to chemoresistance and PDAC cell growth. The PIM kinases are also involved in the PI3K/AKT/mTOR pathway to promote cell survival. In this study, we evaluate the effect of the novel multikinase PIM/PI3K/mTOR inhibitor, AUM302, and commercially available PIM inhibitor, TP-3654. Using five human PDAC cell lines, we found AUM302 to be a potent inhibitor of cell proliferation, cell viability, cell cycle progression, and phosphoprotein expression, while TP-3654 was less effective. Significantly, AUM302 had a strong impact on the viability of gemcitabine-resistant PDAC cells. Taken together, these results demonstrate that AUM302 exhibits antitumor activity in human PDAC cells and thus has the potential to be an effective drug for PDAC therapy.
Collapse
Affiliation(s)
- Komala Ingle
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York, United States of America
| | - Joseph F. LaComb
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York, United States of America
| | - Lee M. Graves
- Department of Pharmacology, School of Medicine, the University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Antonio T. Baines
- Department of Pharmacology, School of Medicine, the University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Biological & Biomedical Sciences, College of Health & Sciences, North Carolina Central University, Durham, North Carolina, United States of America
| | - Agnieszka B. Bialkowska
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York, United States of America
| |
Collapse
|
7
|
Jahankhani K, Ahangari F, Adcock IM, Mortaz E. Possible cancer-causing capacity of COVID-19: Is SARS-CoV-2 an oncogenic agent? Biochimie 2023; 213:130-138. [PMID: 37230238 PMCID: PMC10202899 DOI: 10.1016/j.biochi.2023.05.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 04/24/2023] [Accepted: 05/22/2023] [Indexed: 05/27/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has shown diverse life-threatening effects, most of which are considered short-term. In addition to its short-term effects, which has claimed many millions of lives since 2019, the long-term complications of this virus are still under investigation. Similar to many oncogenic viruses, it has been hypothesized that SARS-CoV-2 employs various strategies to cause cancer in different organs. These include leveraging the renin angiotensin system, altering tumor suppressing pathways by means of its nonstructural proteins, and triggering inflammatory cascades by enhancing cytokine production in the form of a "cytokine storm" paving the way for the emergence of cancer stem cells in target organs. Since infection with SARS-CoV-2 occurs in several organs either directly or indirectly, it is expected that cancer stem cells may develop in multiple organs. Thus, we have reviewed the impact of coronavirus disease 2019 (COVID-19) on the vulnerability and susceptibility of specific organs to cancer development. It is important to note that the cancer-related effects of SARS-CoV-2 proposed in this article are based on the ability of the virus and its proteins to cause cancer but that the long-term consequences of this infection will only be illustrated in the long run.
Collapse
Affiliation(s)
- Kasra Jahankhani
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Ahangari
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ian M Adcock
- Airways Disease, National Heart and Lung Institute, Imperial College London, London, United Kingdom; Immune Health Program at Hunter Medical Research Institute and the College of Health and Medicine at the University of Newcastle, Australia
| | - Esmaeil Mortaz
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Patel MR, Donnellan W, Byrne M, Asch AS, Zeidan AM, Baer MR, Fathi AT, Kuykendall AT, Zheng F, Walker C, Cheng L, Marando C, Savona MR. Phase 1/2 Study of the Pan-PIM Kinase Inhibitor INCB053914 Alone or in Combination With Standard-of-Care Agents in Patients With Advanced Hematologic Malignancies. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2023; 23:674-686. [PMID: 37290996 DOI: 10.1016/j.clml.2023.05.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/28/2023] [Accepted: 05/01/2023] [Indexed: 06/10/2023]
Abstract
BACKGROUND The Proviral Integration site of Moloney murine leukemia virus (PIM) kinases are implicated in tumorigenesis; the pan-PIM kinase inhibitor, INCB053914, demonstrated antitumor activity in hematologic malignancy preclinical models. PATIENTS AND METHODS This phase 1/2 study evaluated oral INCB053914 alone or combined with standard-of-care agents for advanced hematologic malignancies (NCT02587598). In Parts 1/2 (monotherapy), patients (≥18 years) had acute leukemia, high-risk myelodysplastic syndrome (MDS), MDS/myeloproliferative neoplasm, myelofibrosis (MF), multiple myeloma, or lymphoproliferative neoplasms. In Parts 3/4 (combination therapy), patients had relapsed/refractory or newly diagnosed (≥65 years, unfit for intensive chemotherapy) acute myeloid leukemia (AML) or MF with suboptimal ruxolitinib response. RESULTS Parts 1/2 (n = 58): 6 patients experienced dose-limiting toxicities (DLTs), most commonly aspartate aminotransferase/alanine aminotransferase-elevated (AST/ALT; each n = 4). Fifty-seven patients (98.3%) had treatment-emergent adverse events (TEAEs), most commonly ALT-elevated and fatigue (36.2% each); 48 (82.8%) had grade ≥3 TEAEs, most commonly anemia (31.0%); 8 (13.8%) had grade ≥3 ALT/AST-elevated TEAEs. Parts 3/4 (n = 39): for INCB053914 + cytarabine (AML; n = 6), 2 patients experienced DLTs (grade 3 maculopapular rash, n = 1; grade 3 ALT-elevated and grade 4 hypophosphatemia, n = 1); for INCB053914 + azacitidine (AML; n = 16), 1 patient experienced a DLT (grade 3 maculopapular rash). Two complete responses were observed (1 with incomplete count recovery). For INCB053914 + ruxolitinib (MF; n = 17), no DLTs occurred; 3 patients achieved best reduction of >25% spleen volume at week 12 or 24. CONCLUSION INCB053914 was generally well tolerated as monotherapy and in combinations; TEAEs were most commonly ALT/AST-elevated. Limited responses were observed with combinations. Future studies are needed to identify rational, effective combination strategies.
Collapse
Affiliation(s)
- Manish R Patel
- Florida Cancer Specialists/Sarah Cannon Research Institute, Sarasota, FL.
| | | | - Michael Byrne
- Division of Hematology/Oncology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN
| | - Adam S Asch
- Stephenson Cancer Center, Oklahoma University, Oklahoma City, OK
| | - Amer M Zeidan
- Yale University and Yale Cancer Center, New Haven, CT
| | - Maria R Baer
- University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD
| | - Amir T Fathi
- Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | | | | | | | | | | | - Michael R Savona
- Division of Hematology/Oncology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN
| |
Collapse
|
9
|
Deng Z, Richardson DR. The Myc Family and the Metastasis Suppressor NDRG1: Targeting Key Molecular Interactions with Innovative Therapeutics. Pharmacol Rev 2023; 75:1007-1035. [PMID: 37280098 DOI: 10.1124/pharmrev.122.000795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 03/07/2023] [Accepted: 05/01/2023] [Indexed: 06/08/2023] Open
Abstract
Cancer is a leading cause of death worldwide, resulting in ∼10 million deaths in 2020. Major oncogenic effectors are the Myc proto-oncogene family, which consists of three members including c-Myc, N-Myc, and L-Myc. As a pertinent example of the role of the Myc family in tumorigenesis, amplification of MYCN in childhood neuroblastoma strongly correlates with poor patient prognosis. Complexes between Myc oncoproteins and their partners such as hypoxia-inducible factor-1α and Myc-associated protein X (MAX) result in proliferation arrest and pro-proliferative effects, respectively. Interactions with other proteins are also important for N-Myc activity. For instance, the enhancer of zest homolog 2 (EZH2) binds directly to N-Myc to stabilize it by acting as a competitor against the ubiquitin ligase, SCFFBXW7, which prevents proteasomal degradation. Heat shock protein 90 may also be involved in N-Myc stabilization since it binds to EZH2 and prevents its degradation. N-Myc downstream-regulated gene 1 (NDRG1) is downregulated by N-Myc and participates in the regulation of cellular proliferation via associating with other proteins, such as glycogen synthase kinase-3β and low-density lipoprotein receptor-related protein 6. These molecular interactions provide a better understanding of the biologic roles of N-Myc and NDRG1, which can be potentially used as therapeutic targets. In addition to directly targeting these proteins, disrupting their key interactions may also be a promising strategy for anti-cancer drug development. This review examines the interactions between the Myc proteins and other molecules, with a special focus on the relationship between N-Myc and NDRG1 and possible therapeutic interventions. SIGNIFICANCE STATEMENT: Neuroblastoma is one of the most common childhood solid tumors, with a dismal five-year survival rate. This problem makes it imperative to discover new and more effective therapeutics. The molecular interactions between major oncogenic drivers of the Myc family and other key proteins; for example, the metastasis suppressor, NDRG1, may potentially be used as targets for anti-neuroblastoma drug development. In addition to directly targeting these proteins, disrupting their key molecular interactions may also be promising for drug discovery.
Collapse
Affiliation(s)
- Zhao Deng
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Australia (Z.D., D.R.R.), and Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan (D.R.R.)
| | - Des R Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Australia (Z.D., D.R.R.), and Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan (D.R.R.)
| |
Collapse
|
10
|
Hu X, Wu J, Xu J. UCA1 executes an oncogenic role in pancreatic cancer by regulating miR-582-5p/BRCC3. Front Oncol 2023; 13:1133200. [PMID: 37564930 PMCID: PMC10411552 DOI: 10.3389/fonc.2023.1133200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 07/10/2023] [Indexed: 08/12/2023] Open
Abstract
Background As a fatal disease, the mechanism of pancreatic cancer is unclear. Urothelial carcinoma antigen 1(UCA1), a long noncoding RNA (lncRNA) that was first reported in bladder cancer, acts as an oncogene. However, the regulatory role and mechanism of UCA1 in pancreatic cancer remain unknown. This study aims to investigate the expression level and prognostic value of UCA1 in pancreatic cancer tissues, the effects and mechanism of UCA1 in regulating cell proliferation, apoptosis and metastasis. Methods UCA1 expression levels in tissues were detected by in situ hybridization (ISH) and the prognostic value was evaluated by univariate and multivariate survival analysis. For in vitro experiments, proliferation was evaluated by a cell count kit assay, Edu experiments, and a clone formation assay. Apoptosis was evaluated by fluorescence-activated cell sorting flow-cytometry. Cell migration and invasion capacities were detected by wound healing and transwell assays. Western blots were performed to detect apoptotic associated molecules and epithelial-mesenchymal transition (EMT) markers. For the in vivo experiment, subcutaneous transplantation models of pancreatic cancer in nude mice were established to observe the tumor growth. The regulatory mechanism of UCA1 was explored by proteomics, bioinformatic analysis, luciferase reporter assays, and rescue experiments. Results ISH staining revealed that UCA1 levels between cancer tissues (n=94) and tumor-adjacent tissues (n=73) did not show significant differences. Survival analysis indicated that high expression of UCA1 was an unfavorable prognosis factor for pancreatic cancer. Downregulation of UCA1 by siRNA significantly inhibited cell proliferation, decreased the capacities of cell migration and invasion, induced cell apoptosis, and inhibited EMT. Furthermore, we demonstrated that UCA1 positively regulated the expression of BRCC3 by inhibiting miR-582-5p. Rescue experiments indicated that either inhibiting the expression of miR-582-5p or enhancing expression of BRCC3 could partly attenuate the antitumor effects of downregulation of UCA1. Conclusion UCA1 acted as an oncogene in pancreatic cancer by partly regulating miR-582-5p/BRCC3, which could be a new therapeutic target for pancreatic cancer.
Collapse
Affiliation(s)
- Xiaole Hu
- Department of First Operating Room, Qilu Hospital of Shandong University, Jinan, China
| | - Jiahao Wu
- Department of Pancreatic Surgery, General Surgery, Qilu Hospital, Shandong University, Jinan, China
| | - Jianwei Xu
- Department of Pancreatic Surgery, General Surgery, Qilu Hospital, Shandong University, Jinan, China
| |
Collapse
|
11
|
Mansour B, Salem YA, Attallah KM, El-kawy OA, Ibrahim IT, Abdel-Aziz NI. Cyanopyridinone- and Cyanopyridine-Based Cancer Cell Pim-1 Inhibitors: Design, Synthesis, Radiolabeling, Biodistribution, and Molecular Modeling Simulation. ACS OMEGA 2023; 8:19351-19366. [PMID: 37305261 PMCID: PMC10249106 DOI: 10.1021/acsomega.2c08304] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 05/10/2023] [Indexed: 06/13/2023]
Abstract
In this study, two new series of 3-cyanopyridinones (3a-e) and 3-cyanopyridines (4a-e) were synthesized and evaluated for their cytotoxicity and Pim-1 kinase inhibitory activity adopting 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide (MTT) assay and in vitro Pim-1 kinase inhibition assay, respectively. Most of the tested compounds revealed promising cytotoxicity against HepG-2, HCT-116, MCF-7, and PC-3 cell lines. Among them, compounds 4c and 4d showed more potent cytotoxicity against the HePG2 cell line with IC50 = 8.02 ± 0.38 and 6.95 ± 0.34 μM, respectively, than that of the reference 5-FU (IC50 = 9.42 ± 0.46 μM). Moreover, compound 4c was more potent against HCT-116 (IC50 = 7.15 ± 0.35 μM) than 5-FU (IC50 = 8.01 ± 0.39 μM), while compound 4d with IC50 = 8.35 ± 0.42 μM displayed comparable activity to that of the reference drug. Furthermore, high cytotoxic activity was manifested by compounds 4c and 4d against MCF-7 and PC3 cell lines. Our results have also indicated that compounds 4b, 4c, and 4d elicited remarkable inhibition of Pim-1 kinase; 4b and 4c showed equipotent inhibitory activity to that of the reference quercetagetin. Meanwhile, 4d displayed IC50 = 0.46 ± 0.02 μM, showed the best inhibitory activity among the tested compounds, and was more potent than quercetagetin (IC50 = 0.56 ± 0.03 μM). For optimization of the results, docking study of the most potent compounds 4c and 4d in the Pim-1 kinase active site was carried out and compared with both quercetagetin and the reported Pim-1 inhibitor A (VRV), and the results were consistent with those of the biological study. Consequently, compounds 4c and 4d are worthy of further investigations toward the discovery of Pim-1 kinase inhibitors as drug candidates for cancer therapy. Compound 4b was successfully radiolabeled with radioiodine-131, and its biodistribution in Ehrlich ascites carcinoma (EAC)-bearing mice showed more observable uptake in tumor sites, and hence, it can be introduced as a new radiolabeled agent for tumor imaging and therapy.
Collapse
Affiliation(s)
- Basem Mansour
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 11152, Dakahlia, Egypt
| | - Yomna A. Salem
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Sinai University − Kantara Branch, Ismailia 41636, Egypt
| | - Khaled M. Attallah
- Labeled
Compound Department, Hot Lab Center, Egyptian
Atomic Energy Authority, Cairo 13759, Egypt
| | - O. A. El-kawy
- Labeled
Compound Department, Hot Lab Center, Egyptian
Atomic Energy Authority, Cairo 13759, Egypt
| | - Ismail T. Ibrahim
- Labeled
Compound Department, Hot Lab Center, Egyptian
Atomic Energy Authority, Cairo 13759, Egypt
| | - Naglaa I. Abdel-Aziz
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 11152, Dakahlia, Egypt
- Department
of Medicinal Chemistry, Faculty of Pharmacy, University of Mansoura, Mansoura 35516, Egypt
| |
Collapse
|
12
|
Jensen CC, Clements AN, Liou H, Ball LE, Bethard JR, Langlais PR, Toth RK, Chauhan SS, Casillas AL, Daulat SR, Kraft AS, Cress AE, Miranti CK, Mouneimne G, Rogers GC, Warfel NA. PIM1 phosphorylates ABI2 to enhance actin dynamics and promote tumor invasion. J Cell Biol 2023; 222:e202208136. [PMID: 37042842 PMCID: PMC10103708 DOI: 10.1083/jcb.202208136] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 12/21/2022] [Accepted: 03/03/2023] [Indexed: 04/13/2023] Open
Abstract
Distinguishing key factors that drive the switch from indolent to invasive disease will make a significant impact on guiding the treatment of prostate cancer (PCa) patients. Here, we identify a novel signaling pathway linking hypoxia and PIM1 kinase to the actin cytoskeleton and cell motility. An unbiased proteomic screen identified Abl-interactor 2 (ABI2), an integral member of the wave regulatory complex (WRC), as a PIM1 substrate. Phosphorylation of ABI2 at Ser183 by PIM1 increased ABI2 protein levels and enhanced WRC formation, resulting in increased protrusive activity and cell motility. Cell protrusion induced by hypoxia and/or PIM1 was dependent on ABI2. In vivo smooth muscle invasion assays showed that overexpression of PIM1 significantly increased the depth of tumor cell invasion, and treatment with PIM inhibitors significantly reduced intramuscular PCa invasion. This research uncovers a HIF-1-independent signaling axis that is critical for hypoxia-induced invasion and establishes a novel role for PIM1 as a key regulator of the actin cytoskeleton.
Collapse
Affiliation(s)
- Corbin C. Jensen
- Cancer Biology Graduate Program, University of Arizona, Tucson, AZ, USA
| | - Amber N. Clements
- Cancer Biology Graduate Program, University of Arizona, Tucson, AZ, USA
| | - Hope Liou
- Cancer Biology Graduate Program, University of Arizona, Tucson, AZ, USA
| | - Lauren E. Ball
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, USA
| | - Jennifer R. Bethard
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, USA
| | | | | | - Shailender S. Chauhan
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | | | | | | | - Anne E. Cress
- University of Arizona Cancer Center, Tucson, AZ, USA
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Cindy K. Miranti
- University of Arizona Cancer Center, Tucson, AZ, USA
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Ghassan Mouneimne
- University of Arizona Cancer Center, Tucson, AZ, USA
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Greg C. Rogers
- University of Arizona Cancer Center, Tucson, AZ, USA
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Noel A. Warfel
- University of Arizona Cancer Center, Tucson, AZ, USA
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
13
|
Smith PJ, McKeown SR, Patterson LH. Targeting DNA topoisomerase IIα (TOP2A) in the hypoxic tumour microenvironment using unidirectional hypoxia-activated prodrugs (uHAPs). IUBMB Life 2023; 75:40-54. [PMID: 35499745 PMCID: PMC10084299 DOI: 10.1002/iub.2619] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/24/2022] [Accepted: 04/03/2022] [Indexed: 12/29/2022]
Abstract
The hypoxic tumour microenvironment (hTME), arising from inadequate and chaotic vascularity, can present a major obstacle for the treatment of solid tumours. Hypoxic tumour cells compromise responses to treatment since they can generate resistance to radiotherapy, chemotherapy and immunotherapy. The hTME impairs the delivery of a range of anti-cancer drugs, creates routes for metastasis and exerts selection pressures for aggressive phenotypes; these changes potentially occur within an immunosuppressed environment. Therapeutic strategies aimed at the hTME include targeting the molecular changes associated with hypoxia. An alternative approach is to exploit the prevailing lack of oxygen as a principle for the selective activation of prodrugs to target cellular components within the hTME. This review focuses on the design concepts and rationale for the use of unidirectional Hypoxia-Activated Prodrugs (uHAPs) to target the hTME as exemplified by the uHAPs AQ4N and OCT1002. These agents undergo irreversible reduction in a hypoxic environment to active forms that target DNA topoisomerase IIα (TOP2A). This nuclear enzyme is essential for cell division and is a recognised chemotherapeutic target. An activated uHAP interacts with the enzyme-DNA complex to induce DNA damage, cell cycle arrest and tumour cell death. uHAPs are designed to overcome the shortcomings of conventional HAPs and offer unique pharmacodynamic properties for effective targeting of TOP2A in the hTME. uHAP therapy in combination with standard of care treatments has the potential to enhance outcomes by co-addressing the therapeutic challenge presented by the hTME.
Collapse
Affiliation(s)
- Paul J Smith
- Cancer and Genetics Division, School of Medicine, Cardiff University, Cardiff, UK
| | | | - Laurence H Patterson
- Institute of Cancer Therapeutics, School of Pharmacy and Medical Sciences, Faculty of Life Sciences, University of Bradford, Bradford, UK
| |
Collapse
|
14
|
Clements AN, Warfel NA. Targeting PIM Kinases to Improve the Efficacy of Immunotherapy. Cells 2022; 11:3700. [PMID: 36429128 PMCID: PMC9688203 DOI: 10.3390/cells11223700] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/13/2022] [Accepted: 11/16/2022] [Indexed: 11/23/2022] Open
Abstract
The Proviral Integration site for Moloney murine leukemia virus (PIM) kinases is a family of serine/threonine kinases that regulates numerous signaling networks that promote cell growth, proliferation, and survival. PIM kinases are commonly upregulated in both solid tumors and hematological malignancies. Recent studies have demonstrated that PIM facilitates immune evasion in cancer by promoting an immunosuppressive tumor microenvironment that suppresses the innate anti-tumor response. The role of PIM in immune evasion has sparked interest in examining the effect of PIM inhibition in combination with immunotherapy. This review focuses on the role of PIM kinases in regulating immune cell populations, how PIM modulates the immune tumor microenvironment to promote immune evasion, and how PIM inhibitors may be used to enhance the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Amber N. Clements
- Cancer Biology Graduate Program, University of Arizona, Tucson, AZ 85724, USA
| | - Noel A. Warfel
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724, USA
| |
Collapse
|
15
|
Cave DD, Buonaiuto S, Sainz B, Fantuz M, Mangini M, Carrer A, Di Domenico A, Iavazzo TT, Andolfi G, Cortina C, Sevillano M, Heeschen C, Colonna V, Corona M, Cucciardi A, Di Guida M, Batlle E, De Luca A, Lonardo E. LAMC2 marks a tumor-initiating cell population with an aggressive signature in pancreatic cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:315. [PMID: 36289544 PMCID: PMC9609288 DOI: 10.1186/s13046-022-02516-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 10/09/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Tumor-initiating cells (TIC), also known as cancer stem cells, are considered a specific subpopulation of cells necessary for cancer initiation and metastasis; however, the mechanisms by which they acquire metastatic traits are not well understood. METHODS LAMC2 transcriptional levels were evaluated using publicly available transcriptome data sets, and LAMC2 immunohistochemistry was performed using a tissue microarray composed of PDAC and normal pancreas tissues. Silencing and tracing of LAMC2 was performed using lentiviral shRNA constructs and CRISPR/Cas9-mediated homologous recombination, respectively. The contribution of LAMC2 to PDAC tumorigenicity was explored in vitro by tumor cell invasion, migration, sphere-forming and organoids assays, and in vivo by tumor growth and metastatic assays. mRNA sequencing was performed to identify key cellular pathways upregulated in LAMC2 expressing cells. Metastatic spreading induced by LAMC2- expressing cells was blocked by pharmacological inhibition of transforming growth factor beta (TGF-β) signaling. RESULTS We report a LAMC2-expressing cell population, which is endowed with enhanced self-renewal capacity, and is sufficient for tumor initiation and differentiation, and drives metastasis. mRNA profiling of these cells indicates a prominent squamous signature, and differentially activated pathways critical for tumor growth and metastasis, including deregulation of the TGF-β signaling pathway. Treatment with Vactosertib, a new small molecule inhibitor of the TGF-β type I receptor (activin receptor-like kinase-5, ALK5), completely abrogated lung metastasis, primarily originating from LAMC2-expressing cells. CONCLUSIONS We have identified a highly metastatic subpopulation of TICs marked by LAMC2. Strategies aimed at targeting the LAMC2 population may be effective in reducing tumor aggressiveness in PDAC patients. Our results prompt further study of this TIC population in pancreatic cancer and exploration as a potential therapeutic target and/or biomarker.
Collapse
Affiliation(s)
- Donatella Delle Cave
- grid.5326.20000 0001 1940 4177Institute of Genetics and Biophysics “A. Buzzati-Traverso”, National Research Council (CNR-IGB), 80131 Naples, Italy
| | - Silvia Buonaiuto
- grid.5326.20000 0001 1940 4177Institute of Genetics and Biophysics “A. Buzzati-Traverso”, National Research Council (CNR-IGB), 80131 Naples, Italy
| | - Bruno Sainz
- grid.466793.90000 0004 1803 1972Department of Cancer Biology, Instituto de Investigaciones Biomedicas “Alberto Sols” (IIBM), CSIC-UAM, 28029 Madrid, Spain ,grid.420232.50000 0004 7643 3507Chronic Diseases and Cancer, Area 3-Instituto Ramon Y Cajal de Investigacion Sanitaria (IRYCIS), 28034 Madrid, Spain ,grid.510933.d0000 0004 8339 0058Centro de Investigación Biomédica en Red, Área Cáncer, CIBERONC, ISCIII, 28029 Madrid, Spain
| | - Marco Fantuz
- grid.5608.b0000 0004 1757 3470Department of Biology, University of Padova, 35129 Padova, Italy ,grid.428736.cVeneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
| | - Maria Mangini
- grid.5326.20000 0001 1940 4177Institute for Experimental Endocrinology and Oncology, “G. Salvatore” (IEOS), Second Unit, Consiglio Nazionale Delle Ricerche (CNR), 801310 Naples, Italy
| | - Alessandro Carrer
- grid.5608.b0000 0004 1757 3470Department of Biology, University of Padova, 35129 Padova, Italy ,grid.428736.cVeneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
| | - Annalisa Di Domenico
- grid.5326.20000 0001 1940 4177Institute of Genetics and Biophysics “A. Buzzati-Traverso”, National Research Council (CNR-IGB), 80131 Naples, Italy
| | - Tea Teresa Iavazzo
- grid.5326.20000 0001 1940 4177Institute of Genetics and Biophysics “A. Buzzati-Traverso”, National Research Council (CNR-IGB), 80131 Naples, Italy
| | - Gennaro Andolfi
- grid.5326.20000 0001 1940 4177Institute of Genetics and Biophysics “A. Buzzati-Traverso”, National Research Council (CNR-IGB), 80131 Naples, Italy
| | - Carme Cortina
- grid.7722.00000 0001 1811 6966Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, 08028 Barcelona, Spain ,grid.510933.d0000 0004 8339 0058Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 08028 Barcelona, Spain
| | - Marta Sevillano
- grid.7722.00000 0001 1811 6966Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, 08028 Barcelona, Spain ,grid.510933.d0000 0004 8339 0058Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 08028 Barcelona, Spain
| | - Christopher Heeschen
- grid.16821.3c0000 0004 0368 8293State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Vincenza Colonna
- grid.5326.20000 0001 1940 4177Institute of Genetics and Biophysics “A. Buzzati-Traverso”, National Research Council (CNR-IGB), 80131 Naples, Italy
| | - Marco Corona
- grid.5326.20000 0001 1940 4177Institute of Genetics and Biophysics “A. Buzzati-Traverso”, National Research Council (CNR-IGB), 80131 Naples, Italy
| | - Antonio Cucciardi
- grid.5326.20000 0001 1940 4177Institute of Genetics and Biophysics “A. Buzzati-Traverso”, National Research Council (CNR-IGB), 80131 Naples, Italy
| | - Martina Di Guida
- grid.5326.20000 0001 1940 4177Institute of Genetics and Biophysics “A. Buzzati-Traverso”, National Research Council (CNR-IGB), 80131 Naples, Italy
| | - Eduard Batlle
- grid.7722.00000 0001 1811 6966Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, 08028 Barcelona, Spain ,grid.510933.d0000 0004 8339 0058Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 08028 Barcelona, Spain
| | - Annachiara De Luca
- grid.5326.20000 0001 1940 4177Institute for Experimental Endocrinology and Oncology, “G. Salvatore” (IEOS), Second Unit, Consiglio Nazionale Delle Ricerche (CNR), 801310 Naples, Italy
| | - Enza Lonardo
- grid.5326.20000 0001 1940 4177Institute of Genetics and Biophysics “A. Buzzati-Traverso”, National Research Council (CNR-IGB), 80131 Naples, Italy
| |
Collapse
|
16
|
Mahata S, Sahoo PK, Pal R, Sarkar S, Mistry T, Ghosh S, Nasare VD. PIM1/STAT3 axis: a potential co-targeted therapeutic approach in triple-negative breast cancer. Med Oncol 2022; 39:74. [PMID: 35568774 DOI: 10.1007/s12032-022-01675-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 02/01/2022] [Indexed: 10/18/2022]
Abstract
Triple-negative breast cancer lacks an expression of ER, PR, and Her-2, has a poor prognosis, and there are no target therapies available. Therapeutic options to treat TNBC are limited and urgently needed. Strong evidence indicates that molecular signaling pathways have a significant function to regulate biological mechanisms and their abnormal expression endows with the development of cancer. PIM kinase is overexpressed in various human cancers including TNBC which is regulated by various signaling pathways that are crucial for cancer cell proliferation and survival and also make PIM kinase as an attractive drug target. One of the targets of the STAT3 signaling pathway is PIM1 that plays a key role in tumor progression and transformation. In this review, we accumulate the current scenario of the PIM-STAT3 axis that provides insights into the PIM1 and STAT3 inhibitors which can be developed as potential co-inhibitors as prospective anticancer agents.
Collapse
Affiliation(s)
- Sutapa Mahata
- Department of Pathology and Cancer Screening, Chittaranjan National Cancer Institute, 37, S.P. Mukherjee Road, Kolkata, 700026, India
| | - Pranab K Sahoo
- Department of Pathology and Cancer Screening, Chittaranjan National Cancer Institute, 37, S.P. Mukherjee Road, Kolkata, 700026, India
| | - Ranita Pal
- Department of Pathology and Cancer Screening, Chittaranjan National Cancer Institute, 37, S.P. Mukherjee Road, Kolkata, 700026, India
| | - Sinjini Sarkar
- Department of Pathology and Cancer Screening, Chittaranjan National Cancer Institute, 37, S.P. Mukherjee Road, Kolkata, 700026, India
| | - Tanuma Mistry
- Department of Pathology and Cancer Screening, Chittaranjan National Cancer Institute, 37, S.P. Mukherjee Road, Kolkata, 700026, India
| | - Sushmita Ghosh
- Department of Pathology and Cancer Screening, Chittaranjan National Cancer Institute, 37, S.P. Mukherjee Road, Kolkata, 700026, India
| | - Vilas D Nasare
- Department of Pathology and Cancer Screening, Chittaranjan National Cancer Institute, 37, S.P. Mukherjee Road, Kolkata, 700026, India.
| |
Collapse
|
17
|
Popescu VB, Kanhaiya K, Năstac DI, Czeizler E, Petre I. Network controllability solutions for computational drug repurposing using genetic algorithms. Sci Rep 2022; 12:1437. [PMID: 35082323 PMCID: PMC8791995 DOI: 10.1038/s41598-022-05335-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 12/29/2021] [Indexed: 12/22/2022] Open
Abstract
Control theory has seen recently impactful applications in network science, especially in connections with applications in network medicine. A key topic of research is that of finding minimal external interventions that offer control over the dynamics of a given network, a problem known as network controllability. We propose in this article a new solution for this problem based on genetic algorithms. We tailor our solution for applications in computational drug repurposing, seeking to maximize its use of FDA-approved drug targets in a given disease-specific protein-protein interaction network. We demonstrate our algorithm on several cancer networks and on several random networks with their edges distributed according to the Erdős-Rényi, the Scale-Free, and the Small World properties. Overall, we show that our new algorithm is more efficient in identifying relevant drug targets in a disease network, advancing the computational solutions needed for new therapeutic and drug repurposing approaches.
Collapse
Affiliation(s)
| | | | - Dumitru Iulian Năstac
- POLITEHNICA University of Bucharest, Faculty of Electronics, Telecommunications and Information Technology, 061071, Bucharest, Romania
| | - Eugen Czeizler
- Computer Science, Åbo Akademi University, 20500, Turku, Finland
- National Institute for Research and Development in Biological Sciences, 060031, Bucharest, Romania
| | - Ion Petre
- Department of Mathematics and Statistics, University of Turku, 20014, Turku, Finland.
- National Institute for Research and Development in Biological Sciences, 060031, Bucharest, Romania.
| |
Collapse
|
18
|
Zhao Y, Aziz AUR, Zhang H, Zhang Z, Li N, Liu B. A systematic review on active sites and functions of PIM-1 protein. Hum Cell 2022; 35:427-440. [PMID: 35000143 DOI: 10.1007/s13577-021-00656-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 12/02/2021] [Indexed: 12/12/2022]
Abstract
The Proviral Integration of Molony murine leukemia virus (PIM)-1 protein contributes to the solid cancers and hematologic malignancies, cell growth, proliferation, differentiation, migration, and other life activities. Many studies have related these functions to its molecular structure, subcellular localization and expression level. However, recognition of specific active sites and their effects on the activity of this constitutively active kinase is still a challenge. Based on the close relationship between its molecular structure and functional activity, this review covers the specific residues involved in the binding of ATP and different substrates in its catalytic domain. This review then elaborates on the relevant changes in protein conformation and cell functions after PIM-1 binds to different substrates. Therefore, this intensive study can improve the understanding of PIM-1-regulated signaling pathways by facilitating the discovery of its potential phosphorylation substrates.
Collapse
Affiliation(s)
- Youyi Zhao
- School of Biomedical Engineering, Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Dalian, 116024, China
| | - Aziz Ur Rehman Aziz
- School of Biomedical Engineering, Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Dalian, 116024, China
| | - Hangyu Zhang
- School of Biomedical Engineering, Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Dalian, 116024, China
| | - Zhengyao Zhang
- School of Life and Pharmaceutical Sciences, Panjin Campus of Dalian University of Technology, Panjin, 124221, China
| | - Na Li
- School of Biomedical Engineering, Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Dalian, 116024, China.
| | - Bo Liu
- School of Biomedical Engineering, Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Dalian, 116024, China.
| |
Collapse
|
19
|
Relationship between the transcriptional expression of PIM1 and local control in patients with head and neck squamous cell carcinomas treated with radiotherapy. Eur Arch Otorhinolaryngol 2022; 279:3679-3684. [PMID: 34993612 PMCID: PMC9130163 DOI: 10.1007/s00405-021-07223-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 12/13/2021] [Indexed: 11/12/2022]
Abstract
Purpose Proviral integration site for Moloney murine leukemia virus (PIMs) are proto-oncogenes encoding serine/threonine kinases that phosphorylate a variety of substrates involved in the regulation of cellular processes. Elevated expression of PIM-1 has been associated with poor prognosis in several types of cancer. There are no studies that have analyzed the response to radiotherapy in patients with head and neck squamous cell carcinoma (HNSCC) according to the expression of PIM-1. The aim of our study was to analyze the relationship between the transcriptional expression of PIM-1 and local response to radiotherapy in HNSCC patients. Methods We determined the transcriptional expression of PIM-1 in 135 HNSCC patients treated with radiotherapy, including patients treated with chemoradiotherapy (n = 65) and bioradiotherapy (n = 15). Results During the follow-up, 48 patients (35.6%) had a local recurrence of the tumor. Patients with local recurrence had a higher level of PIM-1 expression than those who achieved local control of the disease (P = 0.017). Five-year local recurrence-free survival for patients with a high expression of PIM-1 (n = 43) was 44.6% (95% CI 29.2–60.0%), and for patients with low expression (n = 92) it was 71.9% (95% CI 62.5–81.3%) (P = 0.007). According to the results of multivariate analysis, patients with a high PIM-1 expression had a 2.2-fold increased risk of local recurrence (95% CI 1.22–4.10, P = 0.009). Conclusion Patients with elevated transcriptional expression levels of PIM-1 had a significantly higher risk of local recurrence after radiotherapy. Supplementary Information The online version contains supplementary material available at 10.1007/s00405-021-07223-4.
Collapse
|
20
|
Rathi A, Kumar D, Hasan GM, Haque MM, Hassan MI. Therapeutic targeting of PIM KINASE signaling in cancer therapy: Structural and clinical prospects. Biochim Biophys Acta Gen Subj 2021; 1865:129995. [PMID: 34455019 DOI: 10.1016/j.bbagen.2021.129995] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 07/28/2021] [Accepted: 08/23/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND PIM kinases are well-studied drug targets for cancer, belonging to Serine/Threonine kinases family. They are the downstream target of various signaling pathways, and their up/down-regulation affects various physiological processes. PIM family comprises three isoforms, namely, PIM-1, PIM-2, and PIM-3, on alternative initiation of translation and they have different levels of expression in different types of cancers. Its structure shows a unique ATP-binding site in the hinge region which makes it unique among other kinases. SCOPE OF REVIEW PIM kinases are widely reported in hematological malignancies along with prostate and breast cancers. Currently, many drugs are used as inhibitors of PIM kinases. In this review, we highlighted the physiological significance of PIM kinases in the context of disease progression and therapeutic targeting. We comprehensively reviewed the PIM kinases in terms of their expression and regulation of different physiological roles. We further predicted functional partners of PIM kinases to elucidate their role in the cellular physiology of different cancer and mapped their interaction network. MAJOR CONCLUSIONS A deeper mechanistic insight into the PIM signaling involved in regulating different cellular processes, including transcription, apoptosis, cell cycle regulation, cell proliferation, cell migration and senescence, is provided. Furthermore, structural features of PIM have been dissected to understand the mechanism of inhibition and subsequent implication of designed inhibitors towards therapeutic management of prostate, breast and other cancers. GENERAL SIGNIFICANCE Being a potential drug target for cancer therapy, available drugs and PIM inhibitors at different stages of clinical trials are discussed in detail.
Collapse
Affiliation(s)
- Aanchal Rathi
- Department of Biotechnology, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Dhiraj Kumar
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Gulam Mustafa Hasan
- Department of Biochemistry, College of Medicine, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia
| | | | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India.
| |
Collapse
|
21
|
Challenges for Better Diagnosis and Management of Pancreatic and Biliary Tract Cancers Focusing on Blood Biomarkers: A Systematic Review. Cancers (Basel) 2021; 13:cancers13164220. [PMID: 34439378 PMCID: PMC8394661 DOI: 10.3390/cancers13164220] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Pancreatic and biliary tract cancers are malignant tumors that have a very poor prognosis and are resistant to chemotherapy. The later a cancer is detected, the worse the prognosis becomes; therefore, early detection is important. Biomarkers are physiological indices that serve as a guide to indicate the presence or absence of a certain disease, or its progression. The purpose of our research is to summarize previously reported biomarkers for the diagnosis and prognosis of pancreatic and biliary tract cancers. Abstract Background: pancreatic cancer (PCa) and biliary tract cancer (BTC) are cancers with a poor prognosis and few effective treatments. One of the reasons for this is late detection. Many researchers are tackling to develop non-invasive biomarkers for cancer, but few are specific for PCa or BTC. In addition, genetic abnormalities occur in cancer tissues, which ultimately affect the expression of various molecules. Therefore, it is important to identify molecules that are altered in PCa and BTC. For this systematic review, a systematic review of Medline and Embase to select biomarker studies of PCa and BTC patients was conducted. Results: after reviewing 72 studies, 79 biomarker candidates were identified, including 22 nucleic acids, 43 proteins, and 14 immune cell types. Of the 72 studies, 61 examined PCa, and 11 examined BTC. Conclusion: PCa and BTC are characterized by nucleic acid, protein, and immune cell profiles that are markedly different from those of healthy subjects. These altered molecules and cell subsets may serve as cancer-specific biomarkers, particularly in blood. Further studies are needed to better understand the diagnosis and prognosis of PCa and BTC.
Collapse
|
22
|
Pinelli S, Alinovi R, Poli D, Corradi M, Pelosi G, Tiseo M, Goldoni M, Cavallo D, Mozzoni P. Overexpression of microRNA‑486 affects the proliferation and chemosensitivity of mesothelioma cell lines by targeting PIM1. Int J Mol Med 2021; 47:117. [PMID: 33955505 PMCID: PMC8083808 DOI: 10.3892/ijmm.2021.4950] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 04/08/2021] [Indexed: 12/21/2022] Open
Abstract
Dysregulated levels of microRNAs (miRNAs or miRs), involved in oncogenic pathways, have been proposed to contribute to the aggressiveness of malignant pleural mesothelioma (MPM). Previous studies have highlighted the downregulation of miRNA miR-486-5p in patients with mesothelioma and the introduction of miRNA mimics to restore their reduced or absent functionality in cancer cells is considered an important therapeutic strategy. The aim of the present study was to evaluate the mechanisms through which miRNAs may influence the functions, proliferation and sensitivity to cisplatin of MPM cells. In the present study, a miR-486-5p mimic was transfected into the H2052 and H28 MPM cell lines, and cell viability, proliferation, apoptosis and mitochondrial membrane potential were monitored. miR-486-5p overexpression led to a clear impairment of cell proliferation, targeting CDK4 and attenuating cell cycle progression. In addition, transfection with miR-486-5p mimic negatively regulated the release of inflammatory factors and the expression of Provirus integration site for Moloney murine leukaemia virus 1 (PIM1). The sensitivity of the cells to cisplatin was enhanced by enhancing the apoptotic effects of the drug and impairing mitochondrial function. On the whole, the present study demonstrates that miR-486-5p may play an important role in MPM treatment by targeting multiple pathways involved in tumour development and progression. These activities may be mostly related to the downregulation of PIM1, a crucial regulator of cell survival and proliferation. Furthermore, these results provide support for the combined use of miR-486-5p with chemotherapy as a therapeutic strategy for MPM.
Collapse
Affiliation(s)
- Silvana Pinelli
- Department of Medicine and Surgery, University of Parma, I-43126 Parma, Italy
| | - Rossella Alinovi
- Department of Medicine and Surgery, University of Parma, I-43126 Parma, Italy
| | - Diana Poli
- INAIL Research, Department of Occupational and Environmental Medicine, Epidemiology and Hygiene, Monte Porzio Catone, I-00078 Rome, Italy
| | - Massimo Corradi
- Department of Medicine and Surgery, University of Parma, I-43126 Parma, Italy
| | - Giorgio Pelosi
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, I-43124 Parma, Italy
| | - Marcello Tiseo
- Department of Medicine and Surgery, University of Parma, I-43126 Parma, Italy
| | - Matteo Goldoni
- Department of Medicine and Surgery, University of Parma, I-43126 Parma, Italy
| | - Delia Cavallo
- INAIL Research, Department of Occupational and Environmental Medicine, Epidemiology and Hygiene, Monte Porzio Catone, I-00078 Rome, Italy
| | - Paola Mozzoni
- Department of Medicine and Surgery, University of Parma, I-43126 Parma, Italy
| |
Collapse
|
23
|
Co-Targeting PIM Kinase and PI3K/mTOR in NSCLC. Cancers (Basel) 2021; 13:cancers13092139. [PMID: 33946744 PMCID: PMC8125027 DOI: 10.3390/cancers13092139] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/25/2021] [Accepted: 04/20/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary PIM kinases interact with major oncogenic players, including the PI3K/Akt pathway, and provide an escape mechanism leading to drug resistance. This study examined PIM kinase expression in NSCLC and the potential of PIM1 as a prognostic marker. The effect on cell signaling of novel preclinical PI3K/mTOR/PIM kinase inhibitor IBL-301 was compared to PI3K/mTOR inhibition in vitro and ex vivo. PI3K-mTOR inhibitor sensitive (H1975P) and resistant (H1975GR) cells were compared for altered IL6/STAT3 pathway expression and sensitivity to IBL-301. All three PIM kinases are expressed in NSCLC and PIM1 is a marker of poor prognosis. IBL-301 inhibited c-Myc, the PI3K-Akt and JAK/STAT pathways in vitro and in NSCLC tumor tissue explants. IBL-301 also inhibited secreted pro-inflammatory cytokine MCP-1. PIM kinases were activated in H1975GR cells which were more sensitive to IBL-301 than H1975P cells. A miRNA signature of PI3K-mTOR resistance was validated. Co-targeting PIM kinase and PI3K-mTOR warrants further clinical investigation. Abstract PIM kinases are constitutively active proto-oncogenic serine/threonine kinases that play a role in cell cycle progression, metabolism, inflammation and drug resistance. PIM kinases interact with and stabilize p53, c-Myc and parallel signaling pathway PI3K/Akt. This study evaluated PIM kinase expression in NSCLC and in response to PI3K/mTOR inhibition. It investigated a novel preclinical PI3K/mTOR/PIM inhibitor (IBL-301) in vitro and in patient-derived NSCLC tumor tissues. Western blot analysis confirmed PIM1, PIM2 and PIM3 are expressed in NSCLC cell lines and PIM1 is a marker of poor prognosis in patients with NSCLC. IBL-301 decreased PIM1, c-Myc, pBAD and p4EBP1 (Thr37/46) and peIF4B (S406) protein levels in-vitro and MAP kinase, PI3K-Akt and JAK/STAT pathways in tumor tissue explants. IBL-301 significantly decreased secreted pro-inflammatory cytokine MCP-1. Altered mRNA expression, including activated PIM kinase and c-Myc, was identified in Apitolisib resistant cells (H1975GR) by an IL-6/STAT3 pathway array and validated by Western blot. H1975GR cells were more sensitive to IBL-301 than parent cells. A miRNA array identified a dysregulated miRNA signature of PI3K/mTOR drug resistance consisting of regulators of PIM kinase and c-Myc (miR17-5p, miR19b-3p, miR20a-5p, miR15b-5p, miR203a, miR-206). Our data provides a rationale for co-targeting PIM kinase and PI3K-mTOR to improve therapeutic response in NSCLC.
Collapse
|
24
|
Al-Shaheri FN, Alhamdani MSS, Bauer AS, Giese N, Büchler MW, Hackert T, Hoheisel JD. Blood biomarkers for differential diagnosis and early detection of pancreatic cancer. Cancer Treat Rev 2021; 96:102193. [PMID: 33865174 DOI: 10.1016/j.ctrv.2021.102193] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 03/17/2021] [Accepted: 03/19/2021] [Indexed: 12/12/2022]
Abstract
Pancreatic cancer is currently the most lethal tumor entity and case numbers are rising. It will soon be the second most frequent cause of cancer-related death in the Western world. Mortality is close to incidence and patient survival after diagnosis stands at about five months. Blood-based diagnostics could be one crucial factor for improving this dismal situation and is at a stage that could make this possible. Here, we are reviewing the current state of affairs with its problems and promises, looking at various molecule types. Reported results are evaluated in the overall context. Also, we are proposing steps toward clinical utility that should advance the development toward clinical application by improving biomarker quality but also by defining distinct clinical objectives and the respective diagnostic accuracies required to achieve them. Many of the discussed points and conclusions are highly relevant to other solid tumors, too.
Collapse
Affiliation(s)
- Fawaz N Al-Shaheri
- Division of Functional Genome Analysis, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120 Heidelberg, Germany; Medical Faculty Heidelberg, Heidelberg University, Im Neuenheimer Feld 672, 69120 Heidelberg, Germany.
| | - Mohamed S S Alhamdani
- Division of Functional Genome Analysis, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| | - Andrea S Bauer
- Division of Functional Genome Analysis, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| | - Nathalia Giese
- Department of General Surgery, University Hospital Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany
| | - Markus W Büchler
- Department of General Surgery, University Hospital Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany
| | - Thilo Hackert
- Department of General Surgery, University Hospital Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany
| | - Jörg D Hoheisel
- Division of Functional Genome Analysis, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| |
Collapse
|
25
|
Abstract
IL-6 is involved both in immune responses and in inflammation, hematopoiesis, bone metabolism and embryonic development. IL-6 plays roles in chronic inflammation (closely related to chronic inflammatory diseases, autoimmune diseases and cancer) and even in the cytokine storm of corona virus disease 2019 (COVID-19). Acute inflammation during the immune response and wound healing is a well-controlled response, whereas chronic inflammation and the cytokine storm are uncontrolled inflammatory responses. Non-immune and immune cells, cytokines such as IL-1β, IL-6 and tumor necrosis factor alpha (TNFα) and transcription factors nuclear factor-kappa B (NF-κB) and signal transducer and activator of transcription 3 (STAT3) play central roles in inflammation. Synergistic interactions between NF-κB and STAT3 induce the hyper-activation of NF-κB followed by the production of various inflammatory cytokines. Because IL-6 is an NF-κB target, simultaneous activation of NF-κB and STAT3 in non-immune cells triggers a positive feedback loop of NF-κB activation by the IL-6-STAT3 axis. This positive feedback loop is called the IL-6 amplifier (IL-6 Amp) and is a key player in the local initiation model, which states that local initiators, such as senescence, obesity, stressors, infection, injury and smoking, trigger diseases by promoting interactions between non-immune cells and immune cells. This model counters dogma that holds that autoimmunity and oncogenesis are triggered by the breakdown of tissue-specific immune tolerance and oncogenic mutations, respectively. The IL-6 Amp is activated by a variety of local initiators, demonstrating that the IL-6-STAT3 axis is a critical target for treating diseases.
Collapse
Affiliation(s)
- Toshio Hirano
- National Institutes for Quantum and Radiological Science and Technology, Anagawa, Inage-ku, Chiba, Japan
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
26
|
Saura-Esteller J, Sánchez-Vera I, Núñez-Vázquez S, Jabalquinto-Carrasco J, Cosialls AM, Mendive-Tapia L, Kukhtar D, Martínez-Bueno MD, Lavilla R, Cerón J, Artal-Sanz M, Pons G, Iglesias-Serret D, Gil J. Fluorizoline-induced apoptosis requires prohibitins in nematodes and human cells. Apoptosis 2021; 26:83-95. [PMID: 33387147 DOI: 10.1007/s10495-020-01651-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/11/2020] [Indexed: 02/06/2023]
Abstract
We previously showed that fluorizoline, a fluorinated thiazoline compound, binds to both subunits of the mitochondrial prohibitin (PHB) complex, PHB1 and PHB2, being the expression of these proteins required for fluorizoline-induced apoptosis in mouse embryonic fibroblasts. To investigate the conservation of this apoptotic mechanism, we studied the effect of PHB downregulation on fluorizoline activity on two human cell lines, HEK293T and U2OS. Then, we asked whether PHBs mediate the effect of fluorizoline in a multicellular organism. Interestingly, reduced levels of PHBs in the human cells impaired the induction of apoptosis by fluorizoline. We observed that fluorizoline has a detrimental dose-dependent effect on the development and survival of the nematode model Caenorhabditis elegans. Besides, such effects of fluorizoline treatment in living nematodes were absent in PHB mutants. Finally, we further explored the apoptotic pathway triggered by fluorizoline in human cell lines. We found that the BH3-only proteins NOXA, BIM and PUMA participate in fluorizoline-induced apoptosis and that the induction of NOXA and PUMA is dependent on PHB expression.
Collapse
Affiliation(s)
- José Saura-Esteller
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Ismael Sánchez-Vera
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Sonia Núñez-Vázquez
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Judith Jabalquinto-Carrasco
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Ana M Cosialls
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Lorena Mendive-Tapia
- Laboratory of Medical Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Medicine (IBUB), University of Barcelona, Barcelona, Spain
| | - Dmytro Kukhtar
- Modeling Human Diseases in C. Elegans Group. Genes, Disease and Therapy Program, IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Manuel D Martínez-Bueno
- Andalusian Center for Developmental Biology, Consejo Superior de Investigaciones Científicas/Junta de Andalucía, Universidad Pablo de Olavide, Seville, Spain.,Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, Seville, Spain
| | - Rodolfo Lavilla
- Laboratory of Medical Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Medicine (IBUB), University of Barcelona, Barcelona, Spain
| | - Julián Cerón
- Modeling Human Diseases in C. Elegans Group. Genes, Disease and Therapy Program, IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Marta Artal-Sanz
- Andalusian Center for Developmental Biology, Consejo Superior de Investigaciones Científicas/Junta de Andalucía, Universidad Pablo de Olavide, Seville, Spain.,Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, Seville, Spain
| | - Gabriel Pons
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Daniel Iglesias-Serret
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), L'Hospitalet de Llobregat, Barcelona, Spain.,Facultat de Medicina, Universitat de Vic-Universitat Central de Catalunya (UVic-UCC), Vic, Barcelona, Spain
| | - Joan Gil
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), L'Hospitalet de Llobregat, Barcelona, Spain.
| |
Collapse
|
27
|
Toth RK, Warfel NA. Targeting PIM Kinases to Overcome Therapeutic Resistance in Cancer. Mol Cancer Ther 2020; 20:3-10. [PMID: 33303645 DOI: 10.1158/1535-7163.mct-20-0535] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 08/24/2020] [Accepted: 10/27/2020] [Indexed: 11/16/2022]
Abstract
Cancer progression and the onset of therapeutic resistance are often the results of uncontrolled activation of survival kinases. The proviral integration for the Moloney murine leukemia virus (PIM) kinases are oncogenic serine/threonine kinases that regulate tumorigenesis by phosphorylating a wide range of substrates that control cellular metabolism, proliferation, and survival. Because of their broad impact on cellular processes that facilitate progression and metastasis in many cancer types, it has become clear that the activation of PIM kinases is a significant driver of resistance to various types of anticancer therapies. As a result, efforts to target PIM kinases for anticancer therapy have intensified in recent years. Clinical and preclinical studies indicate that pharmacologic inhibition of PIM has the potential to significantly improve the efficacy of standard and targeted therapies. This review focuses on the signaling pathways through which PIM kinases promote cancer progression and resistance to therapy, as well as highlights biological contexts and promising strategies to exploit PIM as a therapeutic target in cancer.
Collapse
Affiliation(s)
- Rachel K Toth
- University of Arizona Cancer Center, Tucson, Arizona
| | - Noel A Warfel
- University of Arizona Cancer Center, Tucson, Arizona. .,Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona
| |
Collapse
|
28
|
The role of histone methylation in the development of digestive cancers: a potential direction for cancer management. Signal Transduct Target Ther 2020; 5:143. [PMID: 32747629 PMCID: PMC7398912 DOI: 10.1038/s41392-020-00252-1] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 06/22/2020] [Accepted: 07/15/2020] [Indexed: 02/08/2023] Open
Abstract
Digestive cancers are the leading cause of cancer-related death worldwide and have high risks of morbidity and mortality. Histone methylation, which is mediated mainly by lysine methyltransferases, lysine demethylases, and protein arginine methyltransferases, has emerged as an essential mechanism regulating pathological processes in digestive cancers. Under certain conditions, aberrant expression of these modifiers leads to abnormal histone methylation or demethylation in the corresponding cancer-related genes, which contributes to different processes and phenotypes, such as carcinogenesis, proliferation, metabolic reprogramming, epithelial–mesenchymal transition, invasion, and migration, during digestive cancer development. In this review, we focus on the association between histone methylation regulation and the development of digestive cancers, including gastric cancer, liver cancer, pancreatic cancer, and colorectal cancer, as well as on its clinical application prospects, aiming to provide a new perspective on the management of digestive cancers.
Collapse
|
29
|
Michaud DS, Ruan M, Koestler DC, Alonso L, Molina-Montes E, Pei D, Marsit CJ, De Vivo I, Malats N, Kelsey KT. DNA Methylation-Derived Immune Cell Profiles, CpG Markers of Inflammation, and Pancreatic Cancer Risk. Cancer Epidemiol Biomarkers Prev 2020; 29:1577-1585. [PMID: 32430337 DOI: 10.1158/1055-9965.epi-20-0378] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/09/2020] [Accepted: 05/13/2020] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Pancreatic cancer is projected to become the second most common cause of cancer-related death over the next 5 years. Because inflammation is thought to be a common trajectory for disease initiation, we sought to prospectively characterize immune profiles using DNA methylation markers and examine DNA methylation levels previously linked to inflammation biomarkers to evaluate whether these immune markers play a key role in pancreatic cancer. METHODS In a nested case-control study pooling three U.S. prospective cohort studies, DNA methylation was measured in prediagnostic leukocytes of incident pancreatic cancer cases and matched controls using the Illumina MethylationEPIC array. Differentially methylated regions were used to predict immune cell types, and CpGs previously associated with inflammatory biomarkers were selected for the analysis. DNA methylation data from a retrospective case-control study conducted in Spain (PanGenEU) was used for independent replication. RESULTS Immune cell proportions and ratio of cell proportions were not associated with pancreatic cancer risk in the nested case-control study. Methylation extent of CpGs residing in or near gene MNDA was significantly associated with pancreatic cancer risk in the nested case-control study and replicated in PanGenEU. Methylation level of a promoter CpG of gene PIM-1 was associated with survival in both studies. CONCLUSIONS Using a targeted approach, we identified several CpGs that may play a role in pancreatic carcinogenesis in two large, independent studies with distinct study designs. IMPACT These findings could provide insight into critical pathways that may help identify new markers of early disease and survival.
Collapse
Affiliation(s)
- Dominique S Michaud
- Department of Public Health & Community Medicine, Tufts University School of Medicine, Tufts University, Boston, Massachusetts.
- Department of Epidemiology, Brown University, Providence, Rhode Island
| | - Mengyuan Ruan
- Department of Public Health & Community Medicine, Tufts University School of Medicine, Tufts University, Boston, Massachusetts
| | - Devin C Koestler
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, Kansas
- University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, Kansas
| | - Lola Alonso
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO) and CIBERONC, Madrid, Spain
| | - Esther Molina-Montes
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO) and CIBERONC, Madrid, Spain
| | - Dong Pei
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, Kansas
- University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, Kansas
| | - Carmen J Marsit
- Department of Environmental Health and Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Immaculata De Vivo
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Núria Malats
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO) and CIBERONC, Madrid, Spain
| | - Karl T Kelsey
- Department of Epidemiology, Brown University, Providence, Rhode Island
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island
| |
Collapse
|
30
|
Sun Z, Zeng L, Zhang M, Zhang Y, Yang N. PIM1 inhibitor synergizes the anti-tumor effect of osimertinib via STAT3 dephosphorylation in EGFR-mutant non-small cell lung cancer. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:366. [PMID: 32355810 PMCID: PMC7186747 DOI: 10.21037/atm.2020.02.43] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background An increasing amount of evidence has demonstrated that combined or multiple targeted therapies could bring about more durable clinical outcomes, and it is known that epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) resistance is related to bypass activation. This study aims to explore a specific solution for third-generation EGFR-TKI resistance caused by bypass activation, and to examine the antitumor effects of the combination of a novel inhibitor CX-6258 HCl with osimertinib, along with its underlining mechanisms. Methods A bioinformatics analysis was performed to detect the relations between the provirus integration site for Moloney murine leukemia virus 1 (PIM1) expression and prognosis of lung cancer. The EGFR-mutated lung cancer cell lines were treated with the combination of CX-6258 HCl and osimertinib to analyze cell proliferation using the Cell Counting Kit-8, colony formation, and in vivo experiments. Cell migration was analyzed using wound healing and Transwell assays. The apoptosis level was detected using Annexin V-propidium iodide flow cytometry. The expression levels of EGFR and STAT3 were determined using Western blot analysis. Results High expression level of PIM1 was related to the poor prognosis of non-small cell lung cancer (NSCLC). The combined administration of osimertinib and CX-6258 HCl significantly inhibited cell proliferation and migration and effectively induced apoptosis in lung cancer cells. It was more efficient in suppressing EGFR activation and phosphorylation of STAT3 compared with osimertinib treatment alone. Furthermore, it showed a durable efficacy in a xenograft model. Conclusions This study showed that PIM1 is a poor prognostic factor for NSCLC. CX-6258 HCl is a potential molecular inhibitor to sensitize the antitumor effects of osimertinib through the inhibiting of the phosphorylation of STAT3 in NSCLC.
Collapse
Affiliation(s)
- Ziyi Sun
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410006, China.,Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital, and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410006, China
| | - Liang Zeng
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410006, China
| | - Miaomiao Zhang
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410006, China
| | - Yongchang Zhang
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410006, China
| | - Nong Yang
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410006, China
| |
Collapse
|
31
|
Trigg RM, Lee LC, Prokoph N, Jahangiri L, Reynolds CP, Amos Burke GA, Probst NA, Han M, Matthews JD, Lim HK, Manners E, Martinez S, Pastor J, Blanco-Aparicio C, Merkel O, de Los Fayos Alonso IG, Kodajova P, Tangermann S, Högler S, Luo J, Kenner L, Turner SD. The targetable kinase PIM1 drives ALK inhibitor resistance in high-risk neuroblastoma independent of MYCN status. Nat Commun 2019; 10:5428. [PMID: 31780656 PMCID: PMC6883072 DOI: 10.1038/s41467-019-13315-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 11/04/2019] [Indexed: 12/21/2022] Open
Abstract
Resistance to anaplastic lymphoma kinase (ALK)-targeted therapy in ALK-positive non-small cell lung cancer has been reported, with the majority of acquired resistance mechanisms relying on bypass signaling. To proactively identify resistance mechanisms in ALK-positive neuroblastoma (NB), we herein employ genome-wide CRISPR activation screens of NB cell lines treated with brigatinib or ceritinib, identifying PIM1 as a putative resistance gene, whose high expression is associated with high-risk disease and poor survival. Knockdown of PIM1 sensitizes cells of differing MYCN status to ALK inhibitors, and in patient-derived xenografts of high-risk NB harboring ALK mutations, the combination of the ALK inhibitor ceritinib and PIM1 inhibitor AZD1208 shows significantly enhanced anti-tumor efficacy relative to single agents. These data confirm that PIM1 overexpression decreases sensitivity to ALK inhibitors in NB, and suggests that combined front-line inhibition of ALK and PIM1 is a viable strategy for the treatment of ALK-positive NB independent of MYCN status.
Collapse
Affiliation(s)
- Ricky M Trigg
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Lab Block level 3, Box 231, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK.,Functional Genomics, Medicinal Science & Technology, GlaxoSmithKline, Stevenage, SG1 2NY, UK
| | - Liam C Lee
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Lab Block level 3, Box 231, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK.,Amgen, Thousand Oaks, CA, 91320, USA
| | - Nina Prokoph
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Lab Block level 3, Box 231, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| | - Leila Jahangiri
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Lab Block level 3, Box 231, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| | - C Patrick Reynolds
- Cancer Center, Texas Tech University Health Sciences Center School of Medicine, Lubbock, TX, 79430, USA
| | - G A Amos Burke
- Department of Paediatric Oncology, Box 181, Cambridge University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0QQ, UK
| | - Nicola A Probst
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Lab Block level 3, Box 231, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| | - Miaojun Han
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Lab Block level 3, Box 231, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK.,OncoSec, San Diego, CA, 92121, USA
| | - Jamie D Matthews
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Lab Block level 3, Box 231, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| | - Hong Kai Lim
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Lab Block level 3, Box 231, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| | - Eleanor Manners
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Lab Block level 3, Box 231, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| | - Sonia Martinez
- Experimental Therapeutics Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Joaquin Pastor
- Experimental Therapeutics Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Carmen Blanco-Aparicio
- Experimental Therapeutics Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Olaf Merkel
- Department of Experimental Pathology and Laboratory Animal Pathology, Institute of Clinical Pathology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Austria
| | - Ines Garces de Los Fayos Alonso
- Department of Experimental Pathology and Laboratory Animal Pathology, Institute of Clinical Pathology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Austria
| | - Petra Kodajova
- Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, Veterinärplatz 1, Vienna, 1210, Austria
| | - Simone Tangermann
- Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, Veterinärplatz 1, Vienna, 1210, Austria
| | - Sandra Högler
- Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, Veterinärplatz 1, Vienna, 1210, Austria
| | - Ji Luo
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20814, USA
| | - Lukas Kenner
- Department of Experimental Pathology and Laboratory Animal Pathology, Institute of Clinical Pathology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Austria.,Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, Veterinärplatz 1, Vienna, 1210, Austria.,Christian Doppler Laboratory for Applied Metabolomics (CDL-AM), Boltzmanngasse 20, Medical University of Vienna, Vienna, 1090, Austria
| | - Suzanne D Turner
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Lab Block level 3, Box 231, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK.
| |
Collapse
|
32
|
Zhang X, Sun Y, Wang P, Yang C, Li S. Reduced pim-1 expression increases chemotherapeutic drug sensitivity in human androgen-independent prostate cancer cells by inducing apoptosis. Exp Ther Med 2019; 18:2731-2738. [PMID: 31572520 PMCID: PMC6755443 DOI: 10.3892/etm.2019.7862] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 05/16/2019] [Indexed: 12/14/2022] Open
Abstract
Chemotherapeutic drug resistance is an obstacle for the successful therapy of prostate cancer. The aim of the present study was to identify the effects of proto-oncogene serine/threonine-protein kinase pim-1 (pim-1) in the proliferation of chemotherapeutic drug-resistant prostate cancer cells. Androgen-independent human prostate cancer cell lines PC3 and DU145 were used in the current study. Cisplatin-sensitive PC3 cells and cisplatin-resistant PC3/DDP cells were used in drug-resistance assays. The expression levels of pim-1, permeability glycoprotein (p-gp), caspase-3 and cleaved caspase-3 were determined using western blotting analysis; pim-1 was knocked down using pim-1-specific short hairpin RNA (shRNA); cell viability was determined using MTT assay and IC50 values of the chemotherapeutic drugs in human prostate cancer cells tested were calculated using GraphPad 5 software. Androgen-independent human prostate cancer cell lines PC3 and DU145 were transfected with pim-1-targeted or control shRNA, and MTT results revealed that pim-1 knockdown significantly inhibited PC3 and DU145 cell viability in a time-dependent manner (P<0.01). Cisplatin-resistant cells PC3/DDP exhibited higher levels of pim-1 and p-gp expression compared with cisplatin-sensitive PC3 cells; and pim-1 knockdown markedly increased chemotherapeutic drug sensitivity in PC3/DDP cells. In addition, pim-1 knockdown increased chemotherapeutic drug sensitivity in PC3/DDP cells. The molecular mechanism of drug sensitivity was discovered to be partly due to pim-1 knockdown, as it significantly increased apoptosis in cisplatin-resistant PC3/DDP cells. The present study may provide a new strategy for the therapy of prostate cancer.
Collapse
Affiliation(s)
- Xing Zhang
- Department of Urology, Yangzhou Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Yangzhou, Jiangsu 225002, P.R. China
| | - Yuyan Sun
- Department of Urology, Yangzhou Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Yangzhou, Jiangsu 225002, P.R. China
| | - Peng Wang
- Department of Urology, Yangzhou Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Yangzhou, Jiangsu 225002, P.R. China
| | - Changfu Yang
- Department of Urology, Yangzhou Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Yangzhou, Jiangsu 225002, P.R. China
| | - Shengwei Li
- Department of Surgery of Chinese Medicine, Yangzhou Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Yangzhou, Jiangsu 225002, P.R. China
| |
Collapse
|
33
|
Chen J, Tang G. PIM-1 kinase: a potential biomarker of triple-negative breast cancer. Onco Targets Ther 2019; 12:6267-6273. [PMID: 31496730 PMCID: PMC6690594 DOI: 10.2147/ott.s212752] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 07/30/2019] [Indexed: 01/10/2023] Open
Abstract
Triple-negative breast cancer is associated with a poor prognosis, and effective biomarkers for targeted diagnosis and treatment are lacking. The tumorigenicity of the provirus integration site for Moloney murine leukemia virus 1 (PIM-1) gene has been studied for many years. However, its significance in breast cancer remains unclear. In this review we briefly summarized the physiological characteristics and regulation of PIM-1 kinase, and subsequently focused on the role of PIM-1 in tumors, especially breast cancer. Oncogene PIM-1 was found to be upregulated in breast cancer, especially in triple-negative breast cancer. Moreover, it is involved in tumorigenesis and the development of drug resistance, and linked to poor prognosis. A highly selective probe targeting PIM-1 for imaging has emerged, suggesting that PIM-1 may be a potential biomarker for the accurate diagnosis and targeted therapy of triple-negative breast cancer.
Collapse
Affiliation(s)
- Jieying Chen
- Department of Radiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Guangyu Tang
- Department of Radiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
34
|
Gao X, Liu X, Lu Y, Wang Y, Cao W, Liu X, Hu H, Wang H. PIM1 is responsible for IL-6-induced breast cancer cell EMT and stemness via c-myc activation. Breast Cancer 2019; 26:663-671. [PMID: 30989585 PMCID: PMC6694096 DOI: 10.1007/s12282-019-00966-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 03/25/2019] [Indexed: 12/24/2022]
Abstract
Background Interleukin-6 (IL-6) has been demonstrated to be a critical factor for breast cancer malignancy. However, the molecular mechanisms by which IL-6 induce breast cancer cells epithelial–mesenchymal-transition (EMT) and stemness remain elusive. Methods Breast cancer cell lines T47D and MCF7 were exposed to IL-6, the expression of PIM1 was examined by quantitative real-time PCR (qRT-PCR) and western blot. Luciferase reporter assay was used to determine the transcriptional modulation of PIM1 by IL-6 and STAT3 inhibitor. Transwell assay was used to detect the invading ability of breast cancer cells induced by IL-6 or PIM1. The expressions of EMT and stemness markers were determined by qRT-PCR. Results IL-6 promoted PIM1 expression in a dose- and time-dependent manner, and this induction could be abrogated by inhibiting STAT3 activation, subsequently suppressing the transcriptional level of PIM1. Moreover, we noticed that knocking down of PIM1 in cells which was exposed to IL-6 significantly impaired the invasion ability and the expression of EMT and stemness markers. On the contrary, overexpression of PIM1 promoted cell invasion and upregulated the expression of EMT and stemness markers. In addition, we demonstrated that c-myc, the cofactor of PIM1, involved in the pro-oncogenic roles of PIM1. Knocking down of c-myc attenuated the PIM1-induced cell EMT and stemness. Conclusion This study proposed the upregulation of PIM1 by IL-6 contributed to breast cancer cell aggressiveness and targeting PIM1 or c-myc could be novel approaches for breast cancer treatment.
Collapse
Affiliation(s)
- Xueqiang Gao
- Breast Disease Center, The Affiliated Hospital of Qingdao University, 59 Haier Road, Qingdao, 266000, Shandong, China
| | - Xiangping Liu
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
| | - Yangyong Lu
- Department of Galactophore Surgery, Qingdao Women and Children's Hospital, Qingdao, Shandong, 266000, China
| | - Yu Wang
- Breast Disease Center, The Affiliated Hospital of Qingdao University, 59 Haier Road, Qingdao, 266000, Shandong, China
| | - Weihong Cao
- Breast Disease Center, The Affiliated Hospital of Qingdao University, 59 Haier Road, Qingdao, 266000, Shandong, China
| | - Xiaoyi Liu
- Breast Disease Center, The Affiliated Hospital of Qingdao University, 59 Haier Road, Qingdao, 266000, Shandong, China
| | - Haiyan Hu
- Breast Disease Center, The Affiliated Hospital of Qingdao University, 59 Haier Road, Qingdao, 266000, Shandong, China
| | - Haibo Wang
- Breast Disease Center, The Affiliated Hospital of Qingdao University, 59 Haier Road, Qingdao, 266000, Shandong, China.
| |
Collapse
|
35
|
Kuang X, Xiong J, Wang W, Li X, Lu T, Fang Q, Wang J. PIM inhibitor SMI-4a induces cell apoptosis in B-cell acute lymphocytic leukemia cells via the HO-1-mediated JAK2/STAT3 pathway. Life Sci 2019; 219:248-256. [PMID: 30658101 DOI: 10.1016/j.lfs.2019.01.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 01/03/2019] [Accepted: 01/14/2019] [Indexed: 12/14/2022]
Abstract
OBJECTIVES The serine/threonine PIM protein kinases are critical regulators of tumorigenesis in multiple cancers. However, whether PIMs are potential therapeutic targets for treating B-cell acute lymphocytic leukemia (B-ALL) remains unclear. Therefore, here, PIM expression was detected in B-ALL patients and the effects of SMI-4a, a pan-PIM small molecule inhibitor, were investigated in B-ALL cells. METHODS PIM1 and PIM2 expression in 26 newly diagnosed B-ALL cases was detected by real-time PCR and Western blot. B-ALL cells were treated with varied SMI-4a doses and the viability of treated cells was investigated using a cell-counting kit-8 (CCK-8) assay. Apoptosis and cell cycles were analyzed by flow cytometry. Western blot analysis was then used to explore the expression of apoptosis-related proteins and the JAK2/STAT3 pathway. RESULTS PIM1 and 2 were overexpressed in B-ALL patients with high HO-1 level. SMI-4a induced decreases in PIMs and HO-1 expressions and inhibited B-ALL cell viability. Treatment with SMI-4a induced apoptosis by downregulating Bcl-2, upregulating Bax and other antiapoptotic proteins, and decreasing protein levels of p-JAK2 and p-STAT3. In addition, upregulation of HO-1 alleviated decrease in p-JAK2 and p-STAT3 expression, reduced SMI-4a-induced apoptosis of B-ALL cells, and influenced B-ALL cell survival. CONCLUSIONS PIMs were highly expressed in B-ALL patients. SMI-4a inhibited B-ALL proliferation and induced apoptosis via the HO-1-mediated JAK2/STAT3 pathway. SMI-4a might be applicable for treatment of B-ALL cells.
Collapse
Affiliation(s)
- Xingyi Kuang
- Department of Hematology, The Affiliated Hospital of Guizhou Medical University, Guiyang, PR China; Guizhou Province Hematopoietic Stem Cell Transplantation Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, PR China; Key Laboratory of Hematological Disease Diagnostic Treat Centre of Guizhou Province, Guiyang, PR China
| | - Jie Xiong
- Department of Hematology, The Affiliated Hospital of Guizhou Medical University, Guiyang, PR China; Guizhou Province Hematopoietic Stem Cell Transplantation Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, PR China; Key Laboratory of Hematological Disease Diagnostic Treat Centre of Guizhou Province, Guiyang, PR China
| | - Weili Wang
- Department of Hematology, The Affiliated Hospital of Guizhou Medical University, Guiyang, PR China; Guizhou Province Hematopoietic Stem Cell Transplantation Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, PR China; Key Laboratory of Hematological Disease Diagnostic Treat Centre of Guizhou Province, Guiyang, PR China
| | - Xinyao Li
- Department of Hematology, The Affiliated Hospital of Guizhou Medical University, Guiyang, PR China; Guizhou Province Hematopoietic Stem Cell Transplantation Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, PR China; Key Laboratory of Hematological Disease Diagnostic Treat Centre of Guizhou Province, Guiyang, PR China
| | - Tingting Lu
- Department of Hematology, The Affiliated Hospital of Guizhou Medical University, Guiyang, PR China; Guizhou Province Hematopoietic Stem Cell Transplantation Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, PR China; Key Laboratory of Hematological Disease Diagnostic Treat Centre of Guizhou Province, Guiyang, PR China
| | - Qin Fang
- Department of Pharmacy, The Affiliated Baiyun Hospital of Guizhou Medical University, Guiyang, Guizhou, PR China
| | - Jishi Wang
- Department of Hematology, The Affiliated Hospital of Guizhou Medical University, Guiyang, PR China; Guizhou Province Hematopoietic Stem Cell Transplantation Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, PR China; Key Laboratory of Hematological Disease Diagnostic Treat Centre of Guizhou Province, Guiyang, PR China.
| |
Collapse
|
36
|
Zheng H, Yang L, Kang Y, Chen M, Lin S, Xiang Y, Li C, Dai X, Huang X, Liang G, Zhao C. Alantolactone sensitizes human pancreatic cancer cells to EGFR inhibitors through the inhibition of STAT3 signaling. Mol Carcinog 2019; 58:565-576. [PMID: 30520143 DOI: 10.1002/mc.22951] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 11/16/2018] [Accepted: 11/26/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Hailun Zheng
- Chemical Biology Research Center; School of Pharmaceutical Sciences, Wenzhou Medical University; Wenzhou Zhejiang China
| | - Lehe Yang
- Chemical Biology Research Center; School of Pharmaceutical Sciences, Wenzhou Medical University; Wenzhou Zhejiang China
- Division of Pulmonary Medicine; The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung; Wenzhou Zhejiang China
- Department of Respiratory Medicine; Affiliated Yueqing Hospital, Wenzhou Medical University; Wenzhou Zhejiang China
| | - Yanting Kang
- Chemical Biology Research Center; School of Pharmaceutical Sciences, Wenzhou Medical University; Wenzhou Zhejiang China
- Department of Ultrasonography; Yichun People's Hospital; Yichun Jiangxi China
| | - Min Chen
- Chemical Biology Research Center; School of Pharmaceutical Sciences, Wenzhou Medical University; Wenzhou Zhejiang China
| | - Shichong Lin
- Division of Pulmonary Medicine; The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung; Wenzhou Zhejiang China
| | - Youqun Xiang
- Division of Pulmonary Medicine; The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung; Wenzhou Zhejiang China
| | - Caleb Li
- Coffman High School; Dublin Ohio USA
| | - Xuanxuan Dai
- Division of Pulmonary Medicine; The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung; Wenzhou Zhejiang China
| | - Xiaoying Huang
- Division of Pulmonary Medicine; The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung; Wenzhou Zhejiang China
| | - Guang Liang
- Chemical Biology Research Center; School of Pharmaceutical Sciences, Wenzhou Medical University; Wenzhou Zhejiang China
| | - Chengguang Zhao
- Chemical Biology Research Center; School of Pharmaceutical Sciences, Wenzhou Medical University; Wenzhou Zhejiang China
- Department of Respiratory Medicine; Affiliated Yueqing Hospital, Wenzhou Medical University; Wenzhou Zhejiang China
| |
Collapse
|
37
|
Abstract
Pancreatic cancer is a devastating disease with poor prognosis in the modern era. Inflammatory processes have emerged as key mediators of pancreatic cancer development and progression. Recently, studies have been carried out to investigate the underlying mechanisms that contribute to tumorigenesis induced by inflammation. In this review, the role of inflammation in the initiation and progression of pancreatic cancer is discussed.
Collapse
Affiliation(s)
- Kamleshsingh Shadhu
- Pancreas Center of The First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
- Pancreas Institute of Nanjing Medical University, Nanjing, P.R. China
- School of International Education of Nanjing Medical University, Nanjing, P.R. China
| | - Chunhua Xi
- Pancreas Center of The First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
- Pancreas Institute of Nanjing Medical University, Nanjing, P.R. China
| |
Collapse
|
38
|
Taher MY, Davies DM, Maher J. The role of the interleukin (IL)-6/IL-6 receptor axis in cancer. Biochem Soc Trans 2018; 46:1449-1462. [PMID: 30467123 DOI: 10.1042/bst20180136] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 10/19/2018] [Accepted: 10/22/2018] [Indexed: 12/14/2022]
Abstract
Interleukin-6 (IL-6) is a pleiotropic cytokine that activates a classic signalling pathway upon binding to its membrane-bound receptor (IL-6R). Alternatively, IL-6 may 'trans-signal' in a manner that is facilitated by its binding to a soluble derivative of the IL-6 receptor (sIL-6R). Resultant signal transduction is, respectively, driven by the association of IL-6/IL-6R or IL-6/sIL-6R complex with the membrane-associated signal transducer, gp130 (Glycoprotein 130). Distinct JAK (Janus tyrosine kinase)/STAT (signal transducers and activators of transcription) and other signalling pathways are activated as a consequence. Of translational relevance, overexpression of IL-6 has been documented in several neoplastic disorders, including but not limited to colorectal, ovarian and breast cancer and several haematological malignancies. This review attempts to summarise our current understanding of the role of IL-6 in cancer development. In short, these studies have shown important roles for IL-6 signalling in tumour cell growth and survival, angiogenesis, immunomodulation of the tumour microenvironment, stromal cell activation, and ultimate disease progression. Given this background, we also consider the potential for therapeutic targeting of this system in cancer.
Collapse
Affiliation(s)
- Mustafa Yassin Taher
- King's College London, School of Cancer and Pharmaceutical Studies, Guy's Hospital, Great Maze Pond, London SE1 9RT, U.K
- Department of Laboratory Medicine, Taibah University, Medina 42353, Saudi Arabia
| | - David Marc Davies
- King's College London, School of Cancer and Pharmaceutical Studies, Guy's Hospital, Great Maze Pond, London SE1 9RT, U.K
| | - John Maher
- King's College London, School of Cancer and Pharmaceutical Studies, Guy's Hospital, Great Maze Pond, London SE1 9RT, U.K.
- Department of Clinical Immunology and Allergy, King's College Hospital NHS Foundation Trust, London, U.K
- Department of Immunology, Eastbourne Hospital, East Sussex BN21 2UD, U.K
| |
Collapse
|
39
|
Fucic A, Aghajanyan A, Culig Z, Le Novere N. Systems Oncology: Bridging Pancreatic and Castrate Resistant Prostate Cancer. Pathol Oncol Res 2018; 25:1269-1277. [PMID: 30220022 DOI: 10.1007/s12253-018-0467-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 09/03/2018] [Indexed: 12/31/2022]
Abstract
Large investments by pharmaceutical companies in the development of new antineoplastic drugs have not been resulting in adequate advances of new therapies. Despite the introduction of new methods, technologies, translational medicine and bioinformatics, the usage of collected knowledge is unsatisfactory. In this paper, using examples of pancreatic ductal adenocarcinoma (PaC) and castrate-resistant prostate cancer (CRPC), we proposed a concept showing that, in order to improve applicability of current knowledge in oncology, the re-clustering of clinical and scientific data is crucial. Such an approach, based on systems oncology, would include bridging of data on biomarkers and pathways between different cancer types. Proposed concept would introduce a new matrix, which enables combining of already approved therapies between cancer types. Paper provides a (a) detailed analysis of similarities in mechanisms of etiology and progression between PaC and CRPC, (b) diabetes as common hallmark of both cancer types and (c) knowledge gaps and directions of future investigations. Proposed horizontal and vertical matrix in cancer profiling has potency to improve current antineoplastic therapy efficacy. Systems biology map using Systems Biology Graphical Notation Language is used for summarizing complex interactions and similarities of mechanisms in biology of PaC and CRPC.
Collapse
Affiliation(s)
- A Fucic
- Institute for Medical Research and Occupational Health, Ksaverska c 2, 10000, Zagreb, Croatia.
| | - A Aghajanyan
- Institute of Medicine, Peoples' Friendship University of Russia, Moscow, Russian Federation
| | - Z Culig
- Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| | | |
Collapse
|
40
|
Koblish H, Li YL, Shin N, Hall L, Wang Q, Wang K, Covington M, Marando C, Bowman K, Boer J, Burke K, Wynn R, Margulis A, Reuther GW, Lambert QT, Dostalik Roman V, Zhang K, Feng H, Xue CB, Diamond S, Hollis G, Yeleswaram S, Yao W, Huber R, Vaddi K, Scherle P. Preclinical characterization of INCB053914, a novel pan-PIM kinase inhibitor, alone and in combination with anticancer agents, in models of hematologic malignancies. PLoS One 2018; 13:e0199108. [PMID: 29927999 PMCID: PMC6013247 DOI: 10.1371/journal.pone.0199108] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 05/31/2018] [Indexed: 12/17/2022] Open
Abstract
The Proviral Integration site of Moloney murine leukemia virus (PIM) serine/threonine protein kinases are overexpressed in many hematologic and solid tumor malignancies and play central roles in intracellular signaling networks important in tumorigenesis, including the Janus kinase-signal transducer and activator of transcription (JAK/STAT) and phosphatidylinositol 3-kinase (PI3K)/AKT pathways. The three PIM kinase isozymes (PIM1, PIM2, and PIM3) share similar downstream substrates with other key oncogenic kinases and have differing but mutually compensatory functions across tumors. This supports the therapeutic potential of pan-PIM kinase inhibitors, especially in combination with other anticancer agents chosen based on their role in overlapping signaling networks. Reported here is a preclinical characterization of INCB053914, a novel, potent, and selective adenosine triphosphate-competitive pan-PIM kinase inhibitor. In vitro, INCB053914 inhibited proliferation and the phosphorylation of downstream substrates in cell lines from multiple hematologic malignancies. Effects were confirmed in primary bone marrow blasts from patients with acute myeloid leukemia treated ex vivo and in blood samples from patients receiving INCB053914 in an ongoing phase 1 dose-escalation study. In vivo, single-agent INCB053914 inhibited Bcl-2-associated death promoter protein phosphorylation and dose-dependently inhibited tumor growth in acute myeloid leukemia and multiple myeloma xenografts. Additive or synergistic inhibition of tumor growth was observed when INCB053914 was combined with selective PI3Kδ inhibition, selective JAK1 or JAK1/2 inhibition, or cytarabine. Based on these data, pan-PIM kinase inhibitors, including INCB053914, may have therapeutic utility in hematologic malignancies when combined with other inhibitors of oncogenic kinases or standard chemotherapeutics.
Collapse
Affiliation(s)
- Holly Koblish
- Incyte Corporation, Wilmington, Delaware, United States of America
- * E-mail:
| | - Yun-long Li
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Niu Shin
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Leslie Hall
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Qian Wang
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Kathy Wang
- Incyte Corporation, Wilmington, Delaware, United States of America
| | | | - Cindy Marando
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Kevin Bowman
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Jason Boer
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Krista Burke
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Richard Wynn
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Alex Margulis
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Gary W. Reuther
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center, Tampa, Florida, United States of America
| | - Que T. Lambert
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center, Tampa, Florida, United States of America
| | | | - Ke Zhang
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Hao Feng
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Chu-Biao Xue
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Sharon Diamond
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Greg Hollis
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Swamy Yeleswaram
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Wenqing Yao
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Reid Huber
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Kris Vaddi
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Peggy Scherle
- Incyte Corporation, Wilmington, Delaware, United States of America
| |
Collapse
|
41
|
Zhu X, Yu Y, Hou X, Xu J, Tan Z, Nie X, Ling Z, Ge M. Expression of PIM-1 in salivary gland adenoid cystic carcinoma: Association with tumor progression and patients' prognosis. Oncol Lett 2018; 15:1149-1156. [PMID: 29399171 DOI: 10.3892/ol.2017.7408] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 10/13/2017] [Indexed: 01/01/2023] Open
Abstract
Pim-1 proto-oncogene, serine/threonine kinase (PIM-1) phosphorylates a series of substrates to exert its oncogenic function in numerous malignancies. The present study investigated the clinical significance of the PIM-1 protein, apoptosis status and apoptosis-associated proteins, including forkhead box O3a (FOXO3a), B cell lymphoma-2 (BCL-2) and BCL-2-associted agonist of cell death (BAD), were investigated in salivary gland adenoid cystic carcinoma (ACC) tissues. PIM-1 expression levels in 4 pairs of ACC tissues and corresponding normal salivary gland tissues were determined by western blot analysis. PIM-1, FOXO3a, BAD and BCL-2 expression levels in 60 ACC tissues were evaluated by immunohistochemistry (IHC). A terminal deoxynucleotidyl-transferase-mediated dUTP nick end labeling assay was performed to detect the apoptosis status of ACC tissues. PIM-1 was revealed to be highly expressed in ACC tissues compared with adjacent normal tissues. IHC staining results demonstrated high expression ratios of PIM-1, FOXO3a, BCL-2 and BAD [33.33% (20/60), 51.67% (31/60), 51.67% (31/60) and 55% (33/60)], respectively, and significant correlations between the expression of PIM-1 and FOXO3a and BCL-2 (P<0.05). Apoptotic rates were significantly associated with PIM-1, FOXO3a, BCL-2 and BAD expression levels (P<0.05). PIM-1 expression levels were significantly associated with tumor size, lymph node involvement, nerve invasion, distant metastasis and weakly associated with tumor node metastasis stage. Kaplan-Meier survival curves revealed that PIM-1 expression level was significantly associated with disease-free survival of patients with ACC (P=0.009). Cox regression multivariate analysis results revealed that histotype, distant metastasis and apoptotic rate were independent prognosis factors for ACC. Assessment of PIM-1 may be useful in investigating the malignant behaviors of ACC and predicting the outcome of patients with ACC.
Collapse
Affiliation(s)
- Xin Zhu
- Zhejiang Cancer Research Institute, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Yunfang Yu
- Department of Head and Neck Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Xiuxiu Hou
- Department of Head and Neck Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Jiajie Xu
- Department of Head and Neck Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Zhuo Tan
- Department of Head and Neck Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Xilin Nie
- Department of Head and Neck Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Zhiqiang Ling
- Zhejiang Cancer Research Institute, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Minghua Ge
- Department of Head and Neck Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| |
Collapse
|
42
|
Santio NM, Koskinen PJ. PIM kinases: From survival factors to regulators of cell motility. Int J Biochem Cell Biol 2017; 93:74-85. [DOI: 10.1016/j.biocel.2017.10.016] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 10/26/2017] [Accepted: 10/31/2017] [Indexed: 01/01/2023]
|
43
|
Huang PS, Lin YH, Chi HC, Chen PY, Huang YH, Yeh CT, Wang CS, Lin KH. Thyroid hormone inhibits growth of hepatoma cells through induction of miR-214. Sci Rep 2017; 7:14868. [PMID: 29093516 PMCID: PMC5665905 DOI: 10.1038/s41598-017-14864-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 10/05/2017] [Indexed: 12/17/2022] Open
Abstract
Thyroid hormone (TH) plays a role in regulating the metabolic rate, heart functions, muscle control and maintenance of bones. 3,3′5-tri-iodo-L-thyronine (T3) displays high affinity to nuclear thyroid hormone receptors (TRs), which mediate most TH actions. Recent studies have shown hypothyroidism in patients with an increased risk of hepatocellular carcinoma (HCC). MicroRNAs (miRNAs), a class of non-protein-coding RNA, are suggested to control tumor growth by interacting with target genes. However, the clinical significance of T3/TR-regulated miRNAs in tumors has yet to be established. In the current study, miRNA expression profile screening was performed using SYBR Green-Based qRT-PCR array in TR-overexpressing HepG2 cells. miR-214-3p, which is expressed at low levels in HCC, was stimulated upon T3 application. The 3′UTR luciferase reporter assay confirmed that the proto-oncogene serine/threonine-protein kinase, PIM-1, is a miR-214-3p target. PIM-1 was decreased upon treatment with miR-214-3p or T3 stimulation. PIM-1 was highly expressed in HCC, and the effect of PIM-1 on cell proliferation might be mediated by the inhibition of p21. Furthermore, the T3-induced suppression of cell proliferation was partially rescued upon miR-214-3p knockdown. Our data demonstrate that T3 induces miR-214-3p expression and suppresses cell proliferation through PIM-1, thus contributing to the inhibition of HCC tumor formation.
Collapse
Affiliation(s)
- Po-Shuan Huang
- Department of Biochemistry, College of Medicine, Chang-Gung University, 333, Taoyuan, Taiwan
| | - Yang-Hsiang Lin
- Department of Biochemistry, College of Medicine, Chang-Gung University, 333, Taoyuan, Taiwan
| | - Hsiang-Cheng Chi
- Radiation Biology Research Center, Institute for Radiological Research, Chang Gung University/Chang Gung Memorial Hospital, 333, Linkou, Taoyuan, Taiwan
| | - Pei-Yu Chen
- Department of Biochemistry, College of Medicine, Chang-Gung University, 333, Taoyuan, Taiwan
| | - Ya-Hui Huang
- Liver Research Center, Chang Gung Memorial Hospital, 333, Linko, Taoyuan, Taiwan
| | - Chau-Ting Yeh
- Liver Research Center, Chang Gung Memorial Hospital, 333, Linko, Taoyuan, Taiwan
| | - Chia-Siu Wang
- Department of General Surgery, Chang Gung Memorial Hospital, Chiayi, 613, Taiwan.
| | - Kwang-Huei Lin
- Department of Biochemistry, College of Medicine, Chang-Gung University, 333, Taoyuan, Taiwan. .,Liver Research Center, Chang Gung Memorial Hospital, 333, Linko, Taoyuan, Taiwan. .,Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, 333, Taoyuan, Taiwan.
| |
Collapse
|
44
|
Rebello RJ, Huglo AV, Furic L. PIM activity in tumours: A key node of therapy resistance. Adv Biol Regul 2017; 67:163-169. [PMID: 29111105 DOI: 10.1016/j.jbior.2017.10.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 10/20/2017] [Accepted: 10/20/2017] [Indexed: 10/18/2022]
Abstract
The PIM kinases are proto-oncogenes which have been shown to facilitate cell survival and proliferation to drive malignancy and resistance post-therapy. They are able to suppress cell death signals, sustain PI3K/AKT/mTORC1 pathway activity and regulate the MYC oncogenic program. Recent work has revealed PIM kinase essentiality for advanced tumour maintenance and described tumour sensitivity to small molecule inhibitors targeting PIM kinase in multiple malignancies.
Collapse
Affiliation(s)
- Richard J Rebello
- Prostate Cancer Translational Research Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia; Cancer Program, Biomedicine Discovery Institute and Department of Anatomy & Developmental Biology, Monash University, VIC, 3800, Australia
| | - Alisée V Huglo
- Prostate Cancer Translational Research Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Luc Furic
- Prostate Cancer Translational Research Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia; Cancer Program, Biomedicine Discovery Institute and Department of Anatomy & Developmental Biology, Monash University, VIC, 3800, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
45
|
Lui AJ, Geanes ES, Ogony J, Behbod F, Marquess J, Valdez K, Jewell W, Tawfik O, Lewis-Wambi J. IFITM1 suppression blocks proliferation and invasion of aromatase inhibitor-resistant breast cancer in vivo by JAK/STAT-mediated induction of p21. Cancer Lett 2017; 399:29-43. [PMID: 28411130 DOI: 10.1016/j.canlet.2017.04.005] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 03/23/2017] [Accepted: 04/04/2017] [Indexed: 12/19/2022]
Abstract
Interferon induced transmembrane protein 1 (IFITM1) belongs to a family of interferon stimulated genes (ISGs) that is associated with tumor progression and DNA damage resistance; however, its role in endocrine resistance is not known. Here, we correlate IFITM1 expression with clinical stage and poor response to endocrine therapy in a tissue microarray consisting of 94 estrogen receptor (ER)-positive breast tumors. IFITM1 overexpression is confirmed in the AI-resistant MCF-7:5C cell line and not found in AI-sensitive MCF-7 cells. In this study, the orthotopic (mammary fat pad) and mouse mammary intraductal (MIND) models of breast cancer are used to assess tumor growth and invasion in vivo. Lentivirus-mediated shRNA knockdown of IFITM1 in AI-resistant MCF-7:5C cells diminished tumor growth and invasion and induced cell death, whereas overexpression of IFITM1 in wild-type MCF-7 cells promoted estrogen-independent growth and enhanced their aggressive phenotype. Mechanistic studies indicated that loss of IFITM1 in MCF-7:5C cells markedly increased p21 transcription, expression and nuclear localization which was mediated by JAK/STAT activation. These findings suggest IFITM1 overexpression contributes to breast cancer progression and that targeting IFITM1 may be therapeutically beneficial to patients with endocrine-resistant disease.
Collapse
Affiliation(s)
- Asona J Lui
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, USA; The University of Kansas Cancer Center, Kansas City, KS 66160, USA.
| | - Eric S Geanes
- Department of Cancer Biology, University of Kansas Medical Center, USA; The University of Kansas Cancer Center, Kansas City, KS 66160, USA.
| | - Joshua Ogony
- Department of Cancer Biology, University of Kansas Medical Center, USA; The University of Kansas Cancer Center, Kansas City, KS 66160, USA.
| | - Fariba Behbod
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, USA; The University of Kansas Cancer Center, Kansas City, KS 66160, USA.
| | - Jordan Marquess
- University of Kansas Medical Center School of Medicine, USA.
| | - Kelli Valdez
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, USA; The University of Kansas Cancer Center, Kansas City, KS 66160, USA.
| | - William Jewell
- The University of Kansas Cancer Center, Kansas City, KS 66160, USA.
| | - Ossama Tawfik
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, USA.
| | - Joan Lewis-Wambi
- Department of Cancer Biology, University of Kansas Medical Center, USA; The University of Kansas Cancer Center, Kansas City, KS 66160, USA.
| |
Collapse
|
46
|
Pop VV, Seicean A, Lupan I, Samasca G, Burz CC. IL-6 roles - Molecular pathway and clinical implication in pancreatic cancer - A systemic review. Immunol Lett 2017; 181:45-50. [PMID: 27876525 DOI: 10.1016/j.imlet.2016.11.010] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 10/28/2016] [Accepted: 11/18/2016] [Indexed: 02/08/2023]
Abstract
Pancreatic cancer has attracted a great deal of attention owing to the poor outcome, increasing prevalence in the last years and delay diagnosis. Known as a complex disease, it involves genetic mutations, changes in tumour microenvironment and inflammatory component dominated by interleukin-6 and its activated pathways, like Janus Kinase-Signal Transducer and Activator of Translation3, Mitogen Activated Protein Kinase and Androgen receptor. The pro-inflammatory cytokine, plays a central role in oncogenesis, cancer progression, invasiveness, microenvironment changes, treatment resistance, prognosis and associated co morbidities like cachexia and depression. Fulfilling these roles IL-6 requires special attention to understand its complexity in PC development.
Collapse
Affiliation(s)
- Vlad-Vasile Pop
- Iuliu Hatieganu University of Medicine and Pharmacy, Dept. Of Immunology and Allergology, Cluj-Napoca, Romania
| | - Andrada Seicean
- Iuliu Hatieganu University of Medicine and Pharmacy, Dept. Of Internal Medicine, Gastroenterology, Cluj-Napoca, Romania; Octavian Fodor Regional Institute of Gastroenterology and Hepatology, Cluj-Napoca, Romania
| | - Iulia Lupan
- Babes Bolyai University, Department of Molecular Biology, Cluj-Napoca, Romania
| | - Gabriel Samasca
- Iuliu Hatieganu University of Medicine and Pharmacy, Dept. Of Immunology and Allergology, Cluj-Napoca, Romania; Emergency Hospital for Children, Cluj-Napoca, Romania.
| | - Claudia-Cristina Burz
- Iuliu Hatieganu University of Medicine and Pharmacy, Dept. Of Immunology and Allergology, Cluj-Napoca, Romania; Ion Chiricuta Institute of Oncology, Cluj-Napoca, Romania
| |
Collapse
|