1
|
Mokhfi FZ, Al Amin M, Zehravi M, Sweilam SH, Arjun UVNV, Gupta JK, Vallamkonda B, Balakrishnan A, Challa M, Singh J, Prasad PD, Ali SS, Ahmad I, Doukani K, Emran TB. Alkaloid-based modulators of the PI3K/Akt/mTOR pathway for cancer therapy: Understandings from pharmacological point of view. Chem Biol Interact 2024; 402:111218. [PMID: 39209016 DOI: 10.1016/j.cbi.2024.111218] [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: 04/11/2024] [Revised: 06/26/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
This review aims to summarize the role of alkaloids as potential modulators of the PI3K/Akt/mTOR (PAMT) pathway in cancer therapy. The PAMT pathway plays a critical role in cell growth, survival, and metabolism, and its dysregulation contributes to cancer hallmarks. In healthy cells, this pathway is tightly controlled. However, this pathway is frequently dysregulated in cancers and becomes abnormally active. This can happen due to mutations in genes within the pathway itself or due to other factors. This chronic overactivity promotes cancer hallmarks such as uncontrolled cell division, resistance to cell death, and increased blood vessel formation to nourish the tumor. As a result, the PAMT pathway is a crucial therapeutic target for cancer. Researchers are developing drugs that specifically target different components of this pathway, aiming to turn it off and slow cancer progression. Alkaloids, a class of naturally occurring nitrogen-containing molecules found in plants, have emerged as potential therapeutic agents. These alkaloids can target different points within the PAMT pathway, inhibiting its activity and potentially resulting in cancer cell death or suppression of tumor growth. Research is ongoing to explore the role of various alkaloids in cancer treatment. Berberine reduces mTOR activity and increases apoptosis by targeting the PAMT pathway, inhibiting cancer cell proliferation. Lycorine inhibits Akt phosphorylation and mTOR activation, increasing pro-apoptotic protein production and decreasing cell viability. In glioblastoma models, harmine suppresses mTORC1. This review focuses on alkaloids such as evodiamine, hirsuteine, chaetocochin J, indole-3-carbinol, noscapine, berberine, piperlongumine, and so on, which have shown promise in targeting the PAMT pathway. Clinical studies evaluating alkaloids as part of cancer treatment are underway, and their potential impact on patient outcomes is being investigated. In summary, alkaloids represent a promising avenue for targeting the dysregulated PAMT pathway in cancer, and further research is warranted.
Collapse
Affiliation(s)
- Fatima Zohra Mokhfi
- Laboratory of AgroBiotechnology and Nutrition in Semi Arid Zones, Faculty of Nature and Life Sciences, University of Ibn Khaldoun, Tiaret, Algeria
| | - Md Al Amin
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Mehrukh Zehravi
- Department of Clinical Pharmacy, College of Dentistry & Pharmacy, Buraydah Private Colleges, Buraydah, 51418, Saudi Arabia.
| | - Sherouk Hussein Sweilam
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia; Department of Pharmacognosy, Faculty of Pharmacy, Egyptian Russian University, Cairo-Suez Road, Badr City, Cairo, 11829, Egypt
| | - Uppuluri Varuna Naga Venkata Arjun
- Vels Institute of Science, Technology and Advanced Studies (VISTAS), PV Vaithiyalingam Rd, Velan Nagar, Krishna Puram, Pallavaram, Chennai, 600117, Tamil Nadu, India
| | | | - Bhaskar Vallamkonda
- Department of Pharmaceutical Science, School of Applied Sciences and Humanities, VIGNAN'S Foundation for Science, Technology & Research, Vadlamudi, Andhra Pradesh, India
| | - Anitha Balakrishnan
- Department of Pharmaceutics, GRT Institute of Pharmaceutical Education and Research, Tiruttani, Tamil Nadu, India
| | - Manjula Challa
- Department of Pharmaceutics, Vasavi Institute of Pharmaceutical Sciences, Vasavi Nagar, Peddapalli Village, Sidhout Mandal Kadapa District, Andhra Pradesh, India
| | - Jyoti Singh
- School of Pharmaceutical Sciences, Jaipur National University, Jaipur, Rajasthan, India
| | - P Dharani Prasad
- Depertment of Pharmacology, Mohan Babu University, MB School of Pharmaceutical Sciences, (Erstwhile, Sree Vidyaniketan College of Pharmacy), Tirupati, India
| | - Syed Salman Ali
- Lloyd Institute of Management and Technology, Plot No.-11, Knowledge Park-II, Greater Noida, Uttar Pradesh, 201306, India
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Koula Doukani
- Department of Biology, Faculty of Nature and Life Sciences, University of Ibn Khaldoun, Tiaret, Algeria
| | - Talha Bin Emran
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh; Department of Pathology and Laboratory Medicine and Legorreta Cancer Center Warren Alpert Medical School, Brown University, Providence, RI, 02912, USA; Legorreta Cancer Center, Brown University, Providence, RI 02912, USA.
| |
Collapse
|
2
|
Amin M, Gao F, Terrero G, Picus J, Wang-Gillam A, Suresh R, Ma C, Tan B, Baggstrom M, Naughton MJ, Trull L, Belanger S, Fracasso PM, Lockhart AC. Phase I Study of Docetaxel and Temsirolimus in Refractory Solid Tumors. Am J Clin Oncol 2021; 44:443-448. [PMID: 34310349 DOI: 10.1097/coc.0000000000000852] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION The mammalian target of rapamycin (mTOR) is a downstream mediator in the phosphatidylinositol 3-kinase/Akt signaling pathway, and plays a central role in cell proliferation, growth, differentiation, migration, and survival. Temsirolimus (CCI-779), a selective inhibitor of the mTOR, is an ester analog of rapamycin (sirolimus) with improved aqueous solubility and pharmacokinetic (PK) properties. Preclinical studies have confirmed additive and synergistic antitumor activity in cancer cell lines (breast, prostate cancer) with combinations of taxanes and mTOR inhibitors. We conducted a phase I open-label, dose-escalation study to determine the maximal tolerated dose (MTD) of docetaxel in combination with temsirolimus in patients with refractory solid tumors. PATIENTS AND METHODS Eligible patients had a diagnosis of a refractory solid malignancy, measurable disease, and adequate organ function. Patients were sequentially enrolled in 4 dose level intravenous combinations of docetaxel and temsirolimus. Temsirolimus was administered weekly with docetaxel administered every 3 weeks. Laboratory data for tumor markers and radiologic imaging were conducted prestudy and then after every 2 cycles of the treatment. Radiologic response was assessed by Response Evaluation Criteria in Solid Tumors (RECIST) criteria. Blood samples for PK and pharmacodynamic analysis were planned to be drawn at MTD. Apart from the traditional 3+3 design, we also implemented Bayesian Optimal Interval design which uses isotonic regression method to select MTD. We proceeded with isotonic regression analysis by using 20% dose-limiting toxicity (DLT) rate as target. RESULTS Twenty-six patients were treated in this study in 4 cohorts and dose levels. Fourteen males and 12 females were enrolled with a median age of 50 years (range of 27 to 72 y) and median Eastern Cooperative Oncology Group performance score of 1. Tumor histologies included pancreas (6), colon (5), rectum (3), gallbladder (2), non-small cell lung (2), endometrium (1), neuroendocrine (1), esophagus (1), stomach (1), pharynx (1), small intestine (1), and duodenum (1). Stable disease was observed in 2/4 (50%), 3/7 (43%), 4/10 (40%), and 3/5 (60%) patients in cohorts 1, 2, 3, and 4, respectively. Dose escalation in cohorts 2, 3, and 4 was complicated by DLTs such as grade 4 neutropenia and grade 3 diarrhea and an inability for patients to tolerate treatments during and beyond cycle 1 without dose reductions. Therefore, we could not determine an MTD or recommended phase II dose using the traditional 3+3 study analysis. Blood samples for PK and pharmacodynamic analysis were not collected since MTD was not determined. By using 20% DLT rate closest to the target, isotonic regression analysis showed identical estimated DLT rates in dose -1 (docetaxel 50 mg/m2 and temsirolimus 15 mg/m2) and dose level 1 (docetaxel 60mg/m2 and temsirolimus 15 mg/m2). CONCLUSIONS Dose escalation of docetaxel and temsirolimus was limited by severe myelosuppressive toxicity in this phase I study. Most of the DLTs occurred after cycle 1 of therapy hence, we were unable to determine MTD or collect blood samples for PK and pharmacodynamic analysis. Our trial did not meet its objectives due to significant DLTs with this chemotherapy combination. Although our novel use of Bayesian Optimal Interval design using isotonic regression method to select MTD showed identical estimated DLT rates in dose levels 1 and -1, clinically our patients were not able to complete 2 cycles of this regimen without dose reductions due to myelosuppressive toxicity in either of these dose levels, and hence, escaped clinical validity. This combination regimen should not be studied further at the dose levels and schedules tested in our study.
Collapse
Affiliation(s)
- Manik Amin
- Division of Oncology, Dartmouth Hitchcock Medical Center, Lebanon, NH
| | - Feng Gao
- Division of Public Health Sciences, Washington University School of Medicine, Saint Louis, MO
| | - Gretel Terrero
- Division of Hematology/Oncology, Medical University of South Carolina, Hollings Cancer Center, Charleston, SC
| | - Joel Picus
- Division of Oncology, Dartmouth Hitchcock Medical Center, Lebanon, NH
| | | | - Rama Suresh
- Division of Oncology, Dartmouth Hitchcock Medical Center, Lebanon, NH
| | - Cynthia Ma
- Division of Oncology, Dartmouth Hitchcock Medical Center, Lebanon, NH
| | - Benjamin Tan
- Division of Oncology, Dartmouth Hitchcock Medical Center, Lebanon, NH
| | - Maria Baggstrom
- Division of Oncology, Dartmouth Hitchcock Medical Center, Lebanon, NH
| | - Michael J Naughton
- Division of Medical Oncology, Saint Francis Healthcare, Cape Girardeau, MO
| | - Lauren Trull
- Prelude Therapeutics Incorporated, Wilmington, DE
| | - Stephanie Belanger
- Clinical Research Operations at UNC Chapel Hill-Lineberger Comprehensive Cancer Center, Chapel Hill, NC
| | - Paula M Fracasso
- Department of Medicine and the UVA Cancer Center, University of Virginia, Charlottesville, VA
| | - Albert Craig Lockhart
- Division of Hematology/Oncology, Medical University of South Carolina, Hollings Cancer Center, Charleston, SC
| |
Collapse
|
3
|
Hsu JL, Leu WJ, Hsu LC, Ho CH, Liu SP, Guh JH. Phosphodiesterase Type 5 Inhibitors Synergize Vincristine in Killing Castration-Resistant Prostate Cancer Through Amplifying Mitotic Arrest Signaling. Front Oncol 2020; 10:1274. [PMID: 32850387 PMCID: PMC7427565 DOI: 10.3389/fonc.2020.01274] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 06/19/2020] [Indexed: 01/10/2023] Open
Abstract
Combination therapies that display cancer-killing activities through either coexistent targeting of several cellular factors or more efficient suppression of a specific pathway are generally used in cancer treatment. Sildenafil, a specific phosphodiesterase type 5 (PDE5) inhibitor, has been suggested to display both cardioprotective and neuroprotective activities that provide a rationale for the combination with vincristine on the treatment against castration-resistant prostate cancer (CRPC). In the present work, vincristine arrested cells in the metaphase stage of mitosis. Vincristine-induced mitotic arrest was identified by Cdk1 activation (i.e., increased Cdk1Thr161 phosphorylation and decreased Cdk1Tyr15 phosphorylation), cyclin B1 upregulation, and increased phosphorylation of multiple mitotic proteins and stathmin. Sildenafil synergistically potentiated vincristine-induced mitotic arrest and a dramatic increase of mitotic index. Furthermore, sildenafil potentiated vincristine-induced mitochondrial damage, including Mcl-1 downregulation, Bcl-2 phosphorylation and downregulation, Bak upregulation and loss of mitochondrial membrane potential, and sensitized caspase-dependent apoptotic cell death. Sildenafil-mediated synergistic effects were mimicked by other PDE5 inhibitors including vardenafil and tadalafil, and also by PDE5A knockdown in cells, suggesting PDE5-involved mechanism. Notably, sildenafil amplified vincristine-induced phosphorylation and cleavage of BUBR1, a protein kinase in spindle assembly checkpoint (SAC) function and chromosome segregation. Sildenafil also significantly decreased kinetochore tension during SAC activation. Moreover, sildenafil synergized with vincristine on suppressing tumor growth in an in vivo model. In conclusion, the data suggest that sildenafil, in a PDE5-dependent manner, potentiates vincristine-induced mitotic arrest signaling, and sensitizes mitochondria damage–involved apoptosis in CRPC. Both in vitro and in vivo data suggest the combination potential of PDE5 inhibitors and vincristine on CRPC treatment.
Collapse
Affiliation(s)
- Jui-Ling Hsu
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Pharmacy, New Taipei Municipal TuCheng Hospital, Chang Gung Memorial Hospital, New Taipei city, Taiwan
| | - Wohn-Jenn Leu
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Lih-Ching Hsu
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chen-Hsun Ho
- Department of Urology, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan.,Department of Urology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shih-Ping Liu
- Department of Urology, National Taiwan University Hospital College of Medicine, Taipei, Taiwan
| | - Jih-Hwa Guh
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
4
|
Chen K, Shang Z, Dai AL, Dai PL. Novel PI3K/Akt/mTOR pathway inhibitors plus radiotherapy: Strategy for non-small cell lung cancer with mutant RAS gene. Life Sci 2020; 255:117816. [PMID: 32454155 DOI: 10.1016/j.lfs.2020.117816] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/07/2020] [Accepted: 05/16/2020] [Indexed: 02/07/2023]
Abstract
Non-small cell lung cancer (NSCLC) with RAS -mutant gene has been the most difficult obstacle to overcome. Over 25% of muted lung adenocarcinomas have RAS mutation. The prognosis of NSCLC patients with RAS-mutant genes is always poor because there is no effective drug to suppress RAS-mutant genes. NSCLC patients with RAS-mutant usually develop resistance to radiotherapy and chemotherapy, which in some cases leads to a 5-10% survival rate for non-small cell lung cancer (NSCLC). As little clinical symptom of NSCLC was presented at its early stages, thus it always brings in disappointing treatment outcome. Currently, NSCLC presents the highest morbidity and mortality all over the world. The combination of PI3K/AKT/mTOR pathway inhibitors with radiotherapy is a novel strategy to improve radiosensitivity and therapeutic outcome of NSCLC with a RAS-mutant gene. There have been many preclinical studies and clinical trials on the effect of PI3K/AKT/mTOR pathway inhibitors combined with radiotherapy in NSCLC with a RAS-mutant gene have been reported in the past years. This review provides current knowledge of the combination of PI3K/Akt/mTOR pathway inhibitors with radiotherapy, which prove to be a significant improvement for the treatment of NSCLC patients with RAS mutations and will benefit NSCLC patients with RAS mutations.
Collapse
Affiliation(s)
- Kai Chen
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Zhongjun Shang
- Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming 650118, China
| | - Ai-Lin Dai
- Kunming Medical University Haiyuan School, Kunming 650100, China; Maternal and Child Health and Family Planning Service Center of Wenshan state, 663000, China
| | - Pei-Ling Dai
- Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming 650118, China; Kunming Medical University, Kunming 650100, China.
| |
Collapse
|
5
|
Deyell RJ, Wu B, Rassekh SR, Tu D, Samson Y, Fleming A, Bouffet E, Sun X, Powers J, Seymour L, Baruchel S, Morgenstern DA. Phase I study of vinblastine and temsirolimus in pediatric patients with recurrent or refractory solid tumors: Canadian Cancer Trials Group Study IND.218. Pediatr Blood Cancer 2019; 66:e27540. [PMID: 30393943 DOI: 10.1002/pbc.27540] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 09/11/2018] [Accepted: 10/10/2018] [Indexed: 12/26/2022]
Abstract
UNLABELLED Combining mammalian target of rapamycin (mTOR) inhibitors and vinca alkaloids has shown therapeutic synergy in xenograft models of pediatric cancers. This phase I study assessed safety and toxicity of temsirolimus in combination with vinblastine in children. PROCEDURE Patients ≥ 1 and ≤ 18 years with recurrent/refractory solid or CNS tumors were eligible. Vinblastine (4 mg/m2 ) and temsirolimus (15 mg/m2 ) were administered i.v. weekly, with planned dose escalation of vinblastine using a rolling six phase I design. Pharmacokinetic and pharmacodynamic data were collected. RESULTS Seven patients with median age 12 years (range, 8-18 years) were enrolled; all were evaluable for toxicity and six for response. At dose level 1, four of six patients developed grade 3 mucositis, of which one met duration criteria for dose-limiting toxicity (DLT). Four patients required dose omissions for grade 3 or 4 hematologic toxicity, including one prolonged neutropenia DLT. A subsequent patient was enrolled on dose level -2 (temsirolimus 10 mg/m2 , vinblastine 4 mg/m2 ) with no protocol-related toxicity > grade 1, except grade 2 neutropenia. Two serious adverse events (SAE) occurred-an allergic reaction to temsirolimus (grade 2) and an intracranial hemorrhage in a CNS tumor patient (grade 3)-unlikely related to study therapy. Soluble VEGFR2 was reduced at cycle 1, day 36 in keeping with inhibition of angiogenesis. Four patients achieved prolonged stable disease for a median of 5.0 months (range, 3.1-8.3 months). CONCLUSION The combination of weekly temsirolimus (15 mg/m2 ) and vinblastine (4 mg/m2 ) exceeds the maximum tolerated dose in children, with frequent oral mucositis and hematologic toxicity.
Collapse
Affiliation(s)
- Rebecca J Deyell
- Division of Pediatric Hematology/Oncology/BMT, University of British Columbia, British Columbia Children's Hospital and Research Institute, Vancouver, British Columbia, Canada
| | - Bing Wu
- Department of Pediatrics, University of Toronto and New Agent and Innovative Therapy Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - S Rod Rassekh
- Division of Pediatric Hematology/Oncology/BMT, University of British Columbia, British Columbia Children's Hospital and Research Institute, Vancouver, British Columbia, Canada
| | - Dongsheng Tu
- Canadian Cancer Trials Group and Queen's University, Kingston, Ontario, Canada
| | - Yvan Samson
- Centre Hospitalier Universitaire Sainte-Justine, Montreal, Quebec, Canada
| | - Adam Fleming
- McMaster Children's Hospital at Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Eric Bouffet
- Department of Pediatrics, University of Toronto and New Agent and Innovative Therapy Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Xiaoqun Sun
- Canadian Cancer Trials Group and Queen's University, Kingston, Ontario, Canada
| | - Jean Powers
- Canadian Cancer Trials Group and Queen's University, Kingston, Ontario, Canada
| | - Lesley Seymour
- Canadian Cancer Trials Group and Queen's University, Kingston, Ontario, Canada
| | - Sylvain Baruchel
- Department of Pediatrics, University of Toronto and New Agent and Innovative Therapy Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Daniel A Morgenstern
- Department of Pediatrics, University of Toronto and New Agent and Innovative Therapy Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
6
|
Amair-Pinedo F, Matos I, Saurí T, Hernando J, Capdevila J. The Treatment Landscape and New Opportunities of Molecular Targeted Therapies in Gastroenteropancreatic Neuroendocrine Tumors. Target Oncol 2018; 12:757-774. [PMID: 29143176 DOI: 10.1007/s11523-017-0532-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Neuroendocrine neoplasms (NENs) are a heterogeneous group of neoplasms that originate from neuroendocrine stem cells and express both neural and endocrine markers. They are found in almost every organ, and while NENs are mostly associated with slow growth, complications due to the uncontrolled secretion of active peptides, and metastatic disease, may significantly impair the quality of life and can ultimately lead to the death of affected individuals. Expanding knowledge of the genetic, epigenetic, and proteomic landscapes of NENs has led to a better understanding of their molecular pathology and consequently increased treatment options for patients. Here, we review the principal breakthroughs in NEN treatment management, owing largely to omics technologies over the last few years, current recommendations of systemic treatment, and ongoing research into the identification of predictive and response biomarkers based on molecular targeted therapies.
Collapse
Affiliation(s)
| | - Ignacio Matos
- Vall d'Hebron University Hospital, Barcelona, Spain.,Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Tamara Saurí
- Vall d'Hebron University Hospital, Barcelona, Spain.,Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Jorge Hernando
- Vall d'Hebron University Hospital, Barcelona, Spain.,Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Jaume Capdevila
- Vall d'Hebron University Hospital, Barcelona, Spain. .,Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.
| |
Collapse
|
7
|
Phan AT, Dave B. The pivotal role of mammalian target of rapamycin inhibition in the treatment of patients with neuroendocrine tumors. Cancer Med 2016; 5:2953-2964. [PMID: 27539383 PMCID: PMC5083749 DOI: 10.1002/cam4.742] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 12/09/2015] [Accepted: 03/08/2016] [Indexed: 12/31/2022] Open
Abstract
Significant advances have been made toward understanding the biology of neuroendocrine tumors (NET) in terms of defining prognosis and improving clinical management; however, many unmet needs remain. The treatment landscape for NET has evolved, with the approval of the targeted agents everolimus and sunitinib for the treatment of advanced pancreatic NET in 2011 followed by the approval of everolimus for the treatment of advanced nonfunctional gastrointestinal and lung NET in 2016. Mammalian target of rapamycin (mTOR) and components of the mTOR pathway play pivotal roles in NET pathogenesis. Effects of the mTOR inhibitor everolimus have been well documented in preclinical and clinical studies, both as monotherapy and combination therapy. mTOR inhibition as backbone therapy within the NET treatment landscape is a focus of continuing research, which includes evaluation of the growing armamentarium of approved and investigational agents as potential combination partners. Data evaluating the clinical benefits of agents targeting mTOR and related pathways (alone and in combination) in the treatment of patients with NET continue to increase. Many of the findings to date are encouraging.
Collapse
Affiliation(s)
| | - Bhuvanesh Dave
- Houston Methodist Hospital Cancer Center, Houston, Texas
| |
Collapse
|
8
|
|