101
|
Blagden S, Omlin A, Olmin A, Josephs D, Stavraka C, Zivi A, Pinato DJ, Anthoney A, Decordova S, Swales K, Riisnaes R, Pope L, Noguchi K, Shiokawa R, Inatani M, Prince J, Jones K, Twelves C, Spicer J, Banerji U. First-in-human study of CH5132799, an oral class I PI3K inhibitor, studying toxicity, pharmacokinetics, and pharmacodynamics, in patients with metastatic cancer. Clin Cancer Res 2014; 20:5908-17. [PMID: 25231405 DOI: 10.1158/1078-0432.ccr-14-1315] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
PURPOSE This phase I dose-escalation study investigated the maximum-tolerated dose (MTD), dose-limiting toxicities (DLT), safety, pharmacokinetics (PK), pharmacodynamics (PD), and preliminary clinical activity of CH5132799. EXPERIMENTAL DESIGN Patients with metastatic solid tumors were eligible for the study. CH5132799 was administered orally once daily or twice daily in 28-day cycles. RESULTS Thirty-eight patients with solid tumors received CH5132799 at 2 to 96 mg once daily or 48 to 72 mg twice daily. The MTD was 48 mg on the twice-daily schedule but was not reached on the once daily schedule. DLTs were grade 3 elevated liver function tests (LFT), grade 3 fatigue, grade 3 encephalopathy, grade 3 diarrhea, and grade 3 diarrhea with grade 3 stomatitis; all DLTs were reversible. Most drug-related adverse events were grade 1/2. Diarrhea (34%) and nausea (32%) were the most common events. Mean Cmax and AUC0-24 in steady state at MTD were 175 ng/mL and 1,550 ng·h/mL, respectively, consistent with efficacious exposure based on preclinical modeling. Reduction in SUVmax with [(18)F] fluorodeoxyglucose positron emission tomography (FDG-PET) was observed in 5 of 7 patients at MTD. A patient with PIK3CA-mutated clear cell carcinoma of the ovary achieved a partial response by GCIG CA125 criteria and further, a heavily pretreated patient with triple-negative breast cancer had marked improvement in her cutaneous skin lesions lasting six cycles. CONCLUSION CH5132799 is well tolerated at the MTD dose of 48 mg twice daily. At this dose, the drug had a favorable PK and PD profile and preliminary evidence of clinical activity.
Collapse
Affiliation(s)
- Sarah Blagden
- Imperial NIHR/Wellcome Trust Imperial Clinical Research Facility, Imperial College and Hammersmith Hospital Healthcare NHS Trust, London, United Kingdom
| | | | - Aurelius Olmin
- The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Debra Josephs
- King's College London and Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Chara Stavraka
- Imperial NIHR/Wellcome Trust Imperial Clinical Research Facility, Imperial College and Hammersmith Hospital Healthcare NHS Trust, London, United Kingdom
| | - Andrea Zivi
- The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - David J Pinato
- Imperial NIHR/Wellcome Trust Imperial Clinical Research Facility, Imperial College and Hammersmith Hospital Healthcare NHS Trust, London, United Kingdom
| | - Alan Anthoney
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | | | - Karen Swales
- The Institute of Cancer Research, London, United Kingdom
| | - Ruth Riisnaes
- The Institute of Cancer Research, London, United Kingdom
| | - Lorna Pope
- The Institute of Cancer Research, London, United Kingdom
| | | | | | | | - Jenny Prince
- Chugai Pharma Europe Ltd., London, United Kingdom
| | - Keith Jones
- Chugai Pharma Europe Ltd., London, United Kingdom
| | - Chris Twelves
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | - James Spicer
- King's College London and Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Udai Banerji
- The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, United Kingdom.
| |
Collapse
|
102
|
Reijm EA, Timmermans AM, Look MP, Meijer-van Gelder ME, Stobbe CK, van Deurzen CHM, Martens JWM, Sleijfer S, Foekens JA, Berns PMJJ, Jansen MPHM. High protein expression of EZH2 is related to unfavorable outcome to tamoxifen in metastatic breast cancer. Ann Oncol 2014; 25:2185-2190. [PMID: 25193989 DOI: 10.1093/annonc/mdu391] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Metastatic breast cancer (MBC) is a highly heterogeneous disease with great differences in outcome to both chemo- and endocrine therapy. Better insight into the mechanisms underlying resistance is essential to better predict outcome to therapy and to obtain a more tailored treatment approach. We have previously described that increased mRNA expression levels of Enhancer of Zeste homolog (EZH2) are associated with worse outcome to tamoxifen therapy in MBC. Here, we explored whether this is also the case for EZH2 protein expression. PATIENTS AND METHODS A tissue microarray (TMA) was created using formalin-fixed, paraffin-embedded estrogen receptor (ER)-positive primary breast tumor tissues of 250 MBC patients treated with first-line tamoxifen. Quantity and intensity of EZH2 expression were determined by immunohistochemistry (IHC) and both were used to generate and group scores according to a previously described method for scoring EZH2. RESULTS In total, 116 tumors (46%) were considered to be EZH2 positive. The presence of EZH2 protein expression was significantly associated with progression-free survival (PFS) in both univariate [hazard ratio (HR) 1.51, 95% confidence interval (CI) 1.17-1.97, P = 0.002] and multivariate analysis including traditional factors associated with tamoxifen outcome (HR 1.41, 95% CI 1.06-1.88, P = 0.017). Considering quantity irrespective of intensity, tumors with >50% EZH2-positive cells had the worst PFS (HR 2.15, 95% CI 1.42-3.27, P < 0.001), whereas intensity alone did not show a significant association with PFS. Application of other methods of scoring EZH2 positivity resulted in a similar significant association between the amount of EZH2 positive cells and PFS. CONCLUSION In addition to EZH2 mRNA levels, these results suggest that protein expression of EZH2 can be used as a marker to predict outcome to tamoxifen therapy. This provides new rationale to explore EZH2 inhibition in the clinical setting and increases the possibilities for a more personalized treatment approach in MBC patients.
Collapse
Affiliation(s)
- E A Reijm
- Department of Medical Oncology, Cancer Genomics Netherlands
| | - A M Timmermans
- Department of Medical Oncology, Cancer Genomics Netherlands
| | - M P Look
- Department of Medical Oncology, Cancer Genomics Netherlands
| | | | - C K Stobbe
- Department of PATHAN BV, Laboratory Pathology, Sint Franciscus Hospital, Rotterdam, The Netherlands
| | - C H M van Deurzen
- Department of Medical Oncology, Cancer Genomics Netherlands; Department of Pathology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam
| | - J W M Martens
- Department of Medical Oncology, Cancer Genomics Netherlands
| | - S Sleijfer
- Department of Medical Oncology, Cancer Genomics Netherlands
| | - J A Foekens
- Department of Medical Oncology, Cancer Genomics Netherlands
| | - P M J J Berns
- Department of Medical Oncology, Cancer Genomics Netherlands.
| | - M P H M Jansen
- Department of Medical Oncology, Cancer Genomics Netherlands
| |
Collapse
|
103
|
Wozney JL, Antonarakis ES. Growth factor and signaling pathways and their relevance to prostate cancer therapeutics. Cancer Metastasis Rev 2014; 33:581-94. [PMID: 24402967 PMCID: PMC4090293 DOI: 10.1007/s10555-013-9475-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Treatments that target the androgen axis represent an effective strategy for patients with advanced prostate cancer, but the disease remains incurable and new therapeutic approaches are necessary. Significant advances have recently occurred in our understanding of the growth factor and signaling pathways that are active in prostate cancer. In conjunction with this, many new targeted therapies with sound preclinical rationale have entered clinical development and are being tested in men with castration-resistant prostate cancer. Some of the most relevant pathways currently being exploited for therapeutic gain are HGF/c-Met signaling, the PI3K/AKT/mTOR pathway, Hedgehog signaling, the endothelin axis, Src kinase signaling, the IGF pathway, and angiogenesis. Here, we summarize the biological basis for the use of selected targeted agents and the results from available clinical trials of these drugs in men with prostate cancer.
Collapse
Affiliation(s)
- Jocelyn L. Wozney
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - Emmanuel S. Antonarakis
- Prostate Cancer Research Program, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, CRB1-1 M45, 1650 Orleans St., Baltimore, MD 21231, USA
| |
Collapse
|
104
|
Current clinical regulation of PI3K/PTEN/Akt/mTOR signalling in treatment of human cancer. J Cancer Res Clin Oncol 2014; 141:671-89. [PMID: 25146530 DOI: 10.1007/s00432-014-1803-3] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 08/08/2014] [Indexed: 01/14/2023]
Abstract
PURPOSE PTEN is an essential tumour suppressor gene which encodes a phosphatase protein that antagonises the PI3K/Akt/mTOR antiapoptotic pathway. Impairment of this tumour suppressor pathway potentially becomes a causal factor for development of malignancies. This review aims to assess current understanding of mechanisms of dysfunction involving the PI3K/PTEN/Akt/mTOR pathway linked to tumorigenesis and evaluate the evidence for targeted therapy directed at this signalling axis. METHODS Relevant articles in scientific databases were identified using a combination of search terms, including "malignancies", "targeted therapy", "PTEN", and "combination therapy". These databases included Medline, Embase, Cochrane Review, Pubmed, and Scopus. RESULTS PI3K/PTEN expression is frequently deregulated in a majority of malignancies through genetic, epigenetic, and post-transcriptional modifications. This contributes to the upregulation of the PI3K/Akt/mTOR pathway which has been the focus of intense clinical studies. Targeted agents aimed at this pathway offer a novel treatment approach in a variety of haematologic malignancies and solid tumours. Compared to single-agent use, greater response rates were obtained in combination regimens, supporting further investigation of suitable drug combinations in a broad spectrum of malignancies. CONCLUSION Activation of the PI3K/PTEN/Akt/mTOR pathway is implicated both in the pathogenesis of malignancies and development of resistance to anticancer therapies. Therefore, PI3K/Akt/mTOR inhibitors are a promising therapeutic option, in association with systemic cytotoxic and biological therapies, to enable sustained clinical outcomes in cancer treatment. Therapeutic strategies could be tailored according to appropriate biomarkers and patient-specific mutation profiles to maximise benefit of combination therapies.
Collapse
|
105
|
Sheng Y, Li W, Zhu F, Liu K, Chen H, Yao K, Reddy K, Lim DY, Oi N, Li H, Peng C, Ma WY, Bode AM, Dong Z, Dong Z. 3,6,2',4',5'-Pentahydroxyflavone, an orally bioavailable multiple protein kinase inhibitor, overcomes gefitinib resistance in non-small cell lung cancer. J Biol Chem 2014; 289:28192-201. [PMID: 25122774 DOI: 10.1074/jbc.m114.593475] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is the most lethal cancer, causing more than 150,000 deaths in the United States in 2013. The receptor tyrosine kinase inhibitors such as gefitinib are not perfect clinical therapeutic agents for NSCLC treatment due to primary or acquired tyrosine kinase inhibitor resistance. Herein, 3,6,2',4',5'-pentahydroxyflavone (36245-PHF) was identified as a multiple kinase inhibitor for NSCLC treatment based on the computational screening of a natural products database. 36245-PHF was shown to inhibit PI3K and Aurora A and B kinases and overcome gefitinib-resistant NSCLC growth. Our data clearly showed that 36245-PHF markedly inhibited anchorage-independent growth of gefitinib-resistant NSCLC cell lines and exerted a substantial chemotherapeutic effect following oral administration in a gefitinib-resistant NSCLC xenograft model. The evidence from three different subsequent methodological approaches, in vitro, ex vivo, and in vivo, all confirmed that 36245-PHF as a multiple protein kinase inhibitor. Overall, we identified 36245-PHF as a multiple protein kinase inhibitor and as a novel therapeutic agent to overcome gefitinib-resistant NSCLC growth, which could provide a new option for clinical NSCLC oral treatment.
Collapse
Affiliation(s)
- Yuqiao Sheng
- From the The Hormel Institute, University of Minnesota, Austin, Minnesota 55912, the Department of Physiology and Pathophysiology, Basic Medical College, Zhengzhou University, Henan 450001, China, and The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan 450001, China, and
| | - Wei Li
- From the The Hormel Institute, University of Minnesota, Austin, Minnesota 55912, the Cancer Research Institute, Xiangya School of Medicine and
| | - Feng Zhu
- From the The Hormel Institute, University of Minnesota, Austin, Minnesota 55912
| | - Kangdong Liu
- From the The Hormel Institute, University of Minnesota, Austin, Minnesota 55912, the Department of Physiology and Pathophysiology, Basic Medical College, Zhengzhou University, Henan 450001, China, and
| | - Hanyong Chen
- From the The Hormel Institute, University of Minnesota, Austin, Minnesota 55912
| | - Ke Yao
- From the The Hormel Institute, University of Minnesota, Austin, Minnesota 55912
| | - Kanamata Reddy
- From the The Hormel Institute, University of Minnesota, Austin, Minnesota 55912
| | - Do Young Lim
- From the The Hormel Institute, University of Minnesota, Austin, Minnesota 55912
| | - Naomi Oi
- From the The Hormel Institute, University of Minnesota, Austin, Minnesota 55912
| | - Haitao Li
- From the The Hormel Institute, University of Minnesota, Austin, Minnesota 55912
| | - Cong Peng
- From the The Hormel Institute, University of Minnesota, Austin, Minnesota 55912, Xiangya Hospital, Central South University, Changsha, Hunan 410078, China
| | - Wei-Ya Ma
- From the The Hormel Institute, University of Minnesota, Austin, Minnesota 55912
| | - Ann M Bode
- From the The Hormel Institute, University of Minnesota, Austin, Minnesota 55912
| | - Ziming Dong
- the Department of Physiology and Pathophysiology, Basic Medical College, Zhengzhou University, Henan 450001, China, and
| | - Zigang Dong
- From the The Hormel Institute, University of Minnesota, Austin, Minnesota 55912,
| |
Collapse
|
106
|
Fumarola C, Bonelli MA, Petronini PG, Alfieri RR. Targeting PI3K/AKT/mTOR pathway in non small cell lung cancer. Biochem Pharmacol 2014; 90:197-207. [DOI: 10.1016/j.bcp.2014.05.011] [Citation(s) in RCA: 271] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 05/16/2014] [Accepted: 05/16/2014] [Indexed: 01/08/2023]
|
107
|
Paplomata E, O'Regan R. The PI3K/AKT/mTOR pathway in breast cancer: targets, trials and biomarkers. Ther Adv Med Oncol 2014; 6:154-66. [PMID: 25057302 DOI: 10.1177/1758834014530023] [Citation(s) in RCA: 373] [Impact Index Per Article: 33.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The phosphoinositide 3 kinase (PI3K)/Akt/mammalian (or mechanistic) target of rapamycin (mTOR) pathway is a complicated intracellular pathway, which leads to cell growth and tumor proliferation and plays a significant role in endocrine resistance in breast cancer. Multiple compounds targeting this pathway are being evaluated in clinical trials. These agents are generally well tolerated and can be used in combination with targeted therapies, endocrine therapy or cytotoxic agents. The identification of subtypes of tumors more likely to respond to these therapeutics cannot be overemphasized, since breast cancer is a very heterogeneous malignancy. Activation of pathways such as KRAS and MEK can act as escape mechanisms that lead to resistance, thus a combination of agents targeting multiple steps of the intracellular machinery is promising. There is evidence that tumors with PIK3CA mutations are more sensitive to inhibitors of the PI3K pathway but this has yet to be validated. Large clinical trials with correlative studies are necessary to identify reliable biomarkers of efficacy.
Collapse
Affiliation(s)
| | - Ruth O'Regan
- Winship Cancer Institute of Emory University, 1365C Clifton Road, Atlanta, GA 30329, USA
| |
Collapse
|
108
|
Jabbour E, Ottmann OG, Deininger M, Hochhaus A. Targeting the phosphoinositide 3-kinase pathway in hematologic malignancies. Haematologica 2014; 99:7-18. [PMID: 24425689 DOI: 10.3324/haematol.2013.087171] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The phosphoinositide 3-kinase pathway represents an important anticancer target because it has been implicated in cancer cell growth, survival, and motility. Recent studies show that PI3K may also play a role in the development of resistance to currently available therapies. In a broad range of cancers, various components of the phosphoinositide 3-kinase signaling axis are genetically modified, and the pathway can be activated through many different mechanisms. The frequency of genetic alterations in the phosphoinositide 3-kinase pathway, coupled with the impact in oncogenesis and disease progression, make this signaling axis an attractive target in anticancer therapy. A better understanding of the critical function of the phosphoinositide 3-kinase pathway in leukemias and lymphomas has led to the clinical evaluation of novel rationally designed inhibitors in this setting. Three main categories of phosphoinositide 3-kinase inhibitors have been developed so far: agents that target phosphoinositide 3-kinase and mammalian target of rapamycin (dual inhibitors), pan-phosphoinositide 3-kinase inhibitors that target all class I isoforms, and isoform-specific inhibitors that selectively target the α, -β, -γ, or -δ isoforms. Emerging data highlight the promise of phosphoinositide 3-kinase inhibitors in combination with other therapies for the treatment of patients with hematologic malignancies. Further evaluation of phosphoinositide 3-kinase inhibitors in first-line or subsequent regimens may improve clinical outcomes. This article reviews the role of phosphoinositide 3-kinase signaling in hematologic malignancies and the potential clinical utility of inhibitors that target this pathway.
Collapse
|
109
|
Casey SC, Li Y, Fan AC, Felsher DW. Oncogene withdrawal engages the immune system to induce sustained cancer regression. J Immunother Cancer 2014; 2:24. [PMID: 25089198 PMCID: PMC4118610 DOI: 10.1186/2051-1426-2-24] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 06/06/2014] [Indexed: 02/06/2023] Open
Abstract
The targeted inactivation of a single oncogene can induce dramatic tumor regression, suggesting that cancers are “oncogene addicted.” Tumor regression following oncogene inactivation has been thought to be a consequence of restoration of normal physiological programs that induce proliferative arrest, apoptosis, differentiation, and cellular senescence. However, recent observations illustrate that oncogene addiction is highly dependent upon the host immune cells. In particular, CD4+ helper T cells were shown to be essential to the mechanism by which MYC or BCR-ABL inactivation elicits “oncogene withdrawal.” Hence, immune mediators contribute in multiple ways to the pathogenesis, prevention, and treatment of cancer, including mechanisms of tumor initiation, progression, and surveillance, but also oncogene inactivation-mediated tumor regression. Data from both the bench and the bedside illustrates that the inactivation of a driver oncogene can induce activation of the immune system that appears to be essential for sustained tumor regression.
Collapse
Affiliation(s)
- Stephanie C Casey
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, 269 Campus Drive, CCSR 1105, Stanford 94305-5151, CA, USA
| | - Yulin Li
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, 269 Campus Drive, CCSR 1105, Stanford 94305-5151, CA, USA
| | - Alice C Fan
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, 269 Campus Drive, CCSR 1105, Stanford 94305-5151, CA, USA
| | - Dean W Felsher
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, 269 Campus Drive, CCSR 1105, Stanford 94305-5151, CA, USA
| |
Collapse
|
110
|
Targeting the phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) pathway: an emerging treatment strategy for squamous cell lung carcinoma. Cancer Treat Rev 2014; 40:980-9. [PMID: 25037117 DOI: 10.1016/j.ctrv.2014.06.006] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 05/29/2014] [Accepted: 06/08/2014] [Indexed: 01/19/2023]
Abstract
Squamous cell lung carcinoma accounts for approximately 30% of all non-small cell lung cancers (NSCLCs). Despite progress in the understanding of the biology of cancer, cytotoxic chemotherapy remains the standard of care for patients with squamous cell lung carcinoma, but the prognosis is generally poor. The phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) pathway is one of the most commonly activated signaling pathways in cancer, leading to cell proliferation, survival, and differentiation. It has therefore become a major focus of clinical research. Various alterations in the PI3K/AKT/mTOR pathway have been identified in squamous cell lung carcinoma and a number of agents targeting these alterations are in clinical development for use as single agents and in combination with other targeted and conventional treatments. These include pan-PI3K inhibitors, isoform-specific PI3K inhibitors, AKT inhibitors, mTOR inhibitors, and dual PI3K/mTOR inhibitors. These agents have demonstrated antitumor activity in preclinical models of NSCLC and preliminary clinical evidence is also available for some agents. This review will discuss the role of the PI3K/AKT/mTOR pathway in cancer and how the discovery of genetic alterations in this pathway in patients with squamous cell lung carcinoma can inform the development of targeted therapies for this disease. An overview of ongoing clinical trials investigating PI3K/AKT/mTOR pathway inhibitors in squamous cell lung carcinoma will also be included.
Collapse
|
111
|
Vella LJ, Andrews MC, Behren A, Cebon J, Woods K. Immune consequences of kinase inhibitors in development, undergoing clinical trials and in current use in melanoma treatment. Expert Rev Clin Immunol 2014; 10:1107-23. [PMID: 24939732 DOI: 10.1586/1744666x.2014.929943] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Metastatic malignant melanoma is a frequently fatal cancer. In recent years substantial therapeutic progress has occurred with the development of targeted kinase inhibitors and immunotherapeutics. Targeted therapies often result in rapid clinical benefit however responses are seldom durable. Immune therapies can result in durable disease control but responses may not be immediate. Optimal cancer therapy requires both rapid and durable cancer control and this can likely best be achieved by combining targeted therapies with immunotherapeutics. To achieve this, a detailed understanding of the immune consequences of the various kinase inhibitors, in development, clinical trial and currently used to treat melanoma is required.
Collapse
Affiliation(s)
- Laura J Vella
- Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Cancer Immuno-biology Laboratory, Heidelberg, VIC 3084, Australia
| | | | | | | | | |
Collapse
|
112
|
Bowles DW, Senzer N, Hausman D, Peterson S, Vo A, Walker L, Cohen RB, Jimeno A. A multicenter phase 1 study of PX-866 and cetuximab in patients with metastatic colorectal carcinoma or recurrent/metastatic squamous cell carcinoma of the head and neck. Invest New Drugs 2014; 32:1197-203. [PMID: 24916771 DOI: 10.1007/s10637-014-0124-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 06/02/2014] [Indexed: 02/07/2023]
Abstract
BACKGROUND This phase I, dose-finding study determined the safety, maximum tolerated dose (MTD)/recommended phase 2 dose (RP2D), and antitumor activity of PX-866, a phosphatidylinositol 3-kinase inhibitor, combined with cetuximab in patients with incurable colorectal cancer or squamous cell carcinoma of the head and neck. METHODS PX-866 was administered at escalating doses (6-8 mg daily) combined with cetuximab given at a 400 mg/m(2) loading dose followed by 250 mg/m(2) weekly. A "3 + 3" study design was used. Prior therapy with anti-EGFR therapies, including cetuximab, was allowed. RESULTS Eleven patients were enrolled. The most frequent treatment-emergent adverse event was diarrhea (90.1%), followed by hypomagnesemia (72.2%), vomiting (72.2%), fatigue (54.5%), nausea (54.5%), rash (45.5%) and peripheral edema (40%). No dose limiting toxicities were observed. The RP2D was 8 mg, the same as the single-agent PX-866 MTD. Best responses in 9 evaluable patients were: 4 partial responses (44.4%), 4 stable disease (44.4%), and 1 disease progression (11.1%). The median progression free survival was 106 days (range: 1-271). CONCLUSION Treatment with PX-866 and cetuximab was tolerated with signs of anti-tumor activity. Further development of this combination is warranted.
Collapse
MESH Headings
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/blood
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/metabolism
- Cetuximab
- Class I Phosphatidylinositol 3-Kinases
- Colorectal Neoplasms/drug therapy
- Colorectal Neoplasms/genetics
- Colorectal Neoplasms/metabolism
- Disease-Free Survival
- Dose-Response Relationship, Drug
- Female
- Gonanes/administration & dosage
- Gonanes/adverse effects
- Head and Neck Neoplasms/drug therapy
- Head and Neck Neoplasms/genetics
- Head and Neck Neoplasms/metabolism
- Humans
- Male
- Maximum Tolerated Dose
- Middle Aged
- Mutation
- Neoplasm Recurrence, Local/drug therapy
- Neoplasm Recurrence, Local/genetics
- Neoplasm Recurrence, Local/metabolism
- Phosphatidylinositol 3-Kinases/genetics
- Phosphoinositide-3 Kinase Inhibitors
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins p21(ras)
- Response Evaluation Criteria in Solid Tumors
- Squamous Cell Carcinoma of Head and Neck
- ras Proteins/genetics
Collapse
Affiliation(s)
- Daniel W Bowles
- Division of Medical Oncology, University of Colorado School of Medicine, 12801 East 17th Avenue MS 8117 Aurora, Denver, CO, 80045, USA,
| | | | | | | | | | | | | | | |
Collapse
|
113
|
Liu S, Kurzrock R. Toxicity of targeted therapy: Implications for response and impact of genetic polymorphisms. Cancer Treat Rev 2014; 40:883-91. [PMID: 24867380 DOI: 10.1016/j.ctrv.2014.05.003] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2014] [Revised: 05/06/2014] [Accepted: 05/08/2014] [Indexed: 12/11/2022]
Abstract
Targeted therapies have unique toxicity profiles. Common adverse events include rash, diarrhea, hypertension, hypothyroidism, proteinuria, depigmentation, and hepatotoxicity. Some of these toxicities are caused by on-target, mechanism-associated effects, which can be stratified as to whether or not the targets are relevant to response. Other toxicities are off-target and may be caused by the class of agent, e.g. antibody vs small molecule tyrosine kinase inhibitor, or by immune reactions or toxic metabolites. Both on- and off-target toxicities may be due to higher drug concentrations or altered end-organ sensitivity, which in turn can be a consequence of genetic polymorphisms controlling metabolism or tissue responsiveness. On-target toxicities are important to identify as some correlate with response and, hence, amelioration of these side effects is preferable to dose reduction or stopping drug. Toxicities secondary to relevant target impact may be recognized when distinct types of agents, such as antibodies and small molecule kinase inhibitors, with the same target have a similar side effect. For example, both bevacizumab and vascular endothelial growth factor receptor (VEGFR) kinase inhibitors cause hypertension; both epidermal growth factor receptor (EGFR) antibodies and kinase inhibitors cause rash; and these toxicities correlate with response. Herein we review common targeted agent-related toxicities, relevant genetic polymorphisms, and implications for response and patient management.
Collapse
Affiliation(s)
- Sariah Liu
- Division of Hematology and Oncology and Center for Personalized Cancer Therapy, University of California San Diego Moores Cancer Center, United States.
| | - Razelle Kurzrock
- Division of Hematology and Oncology and Center for Personalized Cancer Therapy, University of California San Diego Moores Cancer Center, United States
| |
Collapse
|
114
|
Khan KH, Yap TA, Yan L, Cunningham D. Targeting the PI3K-AKT-mTOR signaling network in cancer. CHINESE JOURNAL OF CANCER 2014; 32:253-65. [PMID: 23642907 PMCID: PMC3845556 DOI: 10.5732/cjc.013.10057] [Citation(s) in RCA: 154] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The phosphoinositide 3-kinase-AKT-mammalian target of rapamycin (PI3K-AKT-mTOR) pathway is a frequently hyperactivated pathway in cancer and is important for tumor cell growth and survival. The development of targeted therapies against mTOR, a vital substrate along this pathway, led to the approval of allosteric inhibitors, including everolimus and temsirolimus, for the treatment of breast, renal, and pancreatic cancers. However, the suboptimal duration of response in unselected patients remains an unresolved issue. Numerous novel therapies against critical nodes of this pathway are therefore being actively investigated in the clinic in multiple tumour types. In this review, we focus on the progress of these agents in clinical development along with their biological rationale, the need of predictive biomarkers and various combination strategies, which will be useful in counteracting the mechanisms of resistance to this class of drugs.
Collapse
Affiliation(s)
- Khurum H Khan
- The Royal Marsden NHS Foundation Trust, Sutton, Surrey, UK
| | | | | | | |
Collapse
|
115
|
Casey SC, Li Y, Felsher DW. An essential role for the immune system in the mechanism of tumor regression following targeted oncogene inactivation. Immunol Res 2014; 58:282-91. [PMID: 24791942 PMCID: PMC4201505 DOI: 10.1007/s12026-014-8503-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Tumors are genetically complex and can have a multitude of mutations. Consequently, it is surprising that the suppression of a single oncogene can result in rapid and sustained tumor regression, illustrating the concept that cancers are often "oncogene addicted." The mechanism of oncogene addiction has been presumed to be largely cell autonomous as a consequence of the restoration of normal physiological programs that induce proliferative arrest, apoptosis, differentiation, and/or cellular senescence. Interestingly, it has recently become apparent that upon oncogene inactivation, the immune response is critical in mediating the phenotypic consequences of oncogene addiction. In particular, CD4(+) T cells have been suggested to be essential to the remodeling of the tumor microenvironment, including the shutdown of host angiogenesis and the induction of cellular senescence in the tumor. However, adaptive and innate immune cells are likely involved. Thus, the effectors of the immune system are involved not only in tumor initiation, tumor progression, and immunosurveillance, but also in the mechanism of tumor regression upon targeted oncogene inactivation. Hence, oncogene inactivation may be an effective therapeutic approach because it both reverses the neoplastic state within a cancer cell and reactivates the host immune response that remodels the tumor microenvironment.
Collapse
Affiliation(s)
- Stephanie C Casey
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, 269 Campus Drive, CCSR 1105, Stanford, CA, 94305-5151, USA
| | | | | |
Collapse
|
116
|
Tsao AS, Roth JA. Novel and Emerging Agents in NSCLC. Lung Cancer 2014. [DOI: 10.1002/9781118468791.ch30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
117
|
Matter MS, Decaens T, Andersen JB, Thorgeirsson SS. Targeting the mTOR pathway in hepatocellular carcinoma: current state and future trends. J Hepatol 2014; 60:855-65. [PMID: 24308993 PMCID: PMC3960348 DOI: 10.1016/j.jhep.2013.11.031] [Citation(s) in RCA: 237] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 10/28/2013] [Accepted: 11/27/2013] [Indexed: 02/07/2023]
Abstract
Mechanistic target of rapamycin (mTOR) regulates cell growth, metabolism and aging in response to nutrients, cellular energy stage and growth factors. mTOR is frequently up-regulated in cancer including hepatocellular carcinoma (HCC) and is associated with bad prognosis, poorly differentiated tumors, and earlier recurrence. Blocking mTOR with rapamycin and first generation mTOR inhibitors, called rapalogs, has shown promising reduction of HCC tumor growth in preclinical models. Currently, rapamycin/rapalogs are used in several clinical trials for the treatment of advanced HCC, and as adjuvant therapy in HCC patients after liver transplantation and TACE. A second generation of mTOR pathway inhibitors has been developed recently and is being tested in various clinical trials of solid cancers, and has been used in preclinical HCC models. The results of series of clinical trials using mTOR inhibitors in HCC treatment will emerge in the near future.
Collapse
|
118
|
Bowles DW, Diamond JR, Lam ET, Weekes CD, Astling DP, Anderson RT, Leong S, Gore L, Varella-Garcia M, Vogler BW, Keysar SB, Freas E, Aisner DL, Ren C, Tan AC, Wilhelm F, Maniar M, Eckhardt SG, Messersmith WA, Jimeno A. Phase I study of oral rigosertib (ON 01910.Na), a dual inhibitor of the PI3K and Plk1 pathways, in adult patients with advanced solid malignancies. Clin Cancer Res 2014; 20:1656-65. [PMID: 24493827 PMCID: PMC4160109 DOI: 10.1158/1078-0432.ccr-13-2506] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
PURPOSE To determine the pharmacokinetics (PK), maximum tolerated dose (MTD), safety, and antitumor activity of an oral formulation of rigosertib, a dual phosphoinositide 3-kinase (PI3K) and polo-like kinase 1 (Plk1) pathway inhibitor, in patients with advanced solid malignancies. EXPERIMENTAL DESIGN Patients with advanced solid malignancies received rigosertib twice daily continuously in 21-day cycles. Doses were escalated until intolerable grade ≥2 toxicities, at which point the previous dose level was expanded to define the MTD. All patients were assessed for safety, PK, and response. Urinary PK were performed at the MTD. Archival tumors were assessed for potential molecular biomarkers with multiplex mutation testing. A subset of squamous cell carcinomas (SCC) underwent exome sequencing. RESULTS Forty-eight patients received a median of 2 cycles of therapy at 5 dose levels. Rigosertib exposure increased with escalating doses. Dose-limiting toxicities were hematuria and dysuria. The most common grade ≥2 drug-related toxicities involved urothelial irritation. The MTD is 560 mg twice daily. Activity was seen in head and neck SCCs (1 complete response, 1 partial response) and stable disease for ≥12 weeks was observed in 8 additional patients. Tumors experiencing ≥partial response had PI3K pathway activation, inactivated p53, and unique variants in ROBO3 and FAT1, two genes interacting with the Wnt/β-catenin pathway. CONCLUSIONS The recommended phase II dose of oral rigosertib is 560 mg twice daily given continuously. Urinary toxicity is the dose-limiting and most common toxicity. Alterations in PI3K, p53, and Wnt/β-catenin pathway signaling should be investigated as potential biomarkers of response in future trials.
Collapse
Affiliation(s)
| | | | - Elaine T. Lam
- Department of Medicine, Division of Medical Oncology
| | | | | | | | - Stephen Leong
- Department of Medicine, Division of Medical Oncology
| | - Lia Gore
- Department of Medicine, Division of Medical Oncology
| | | | | | | | | | - Dara L. Aisner
- Department of Pathology, University of Colorado School of Medicine, Aurora, Colorado
| | - Chen Ren
- Onconova Therapeutics Inc, Newtown, Pennsylvania
| | - Aik-Chook Tan
- Department of Medicine, Division of Medical Oncology
| | | | - Manoj Maniar
- Onconova Therapeutics Inc, Newtown, Pennsylvania
| | | | | | | |
Collapse
|
119
|
Phase I Safety, Pharmacokinetic, and Pharmacodynamic Study of SAR245409 (XL765), a Novel, Orally Administered PI3K/mTOR Inhibitor in Patients with Advanced Solid Tumors. Clin Cancer Res 2014; 20:2445-56. [DOI: 10.1158/1078-0432.ccr-13-2403] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
120
|
Ando Y, Inada-Inoue M, Mitsuma A, Yoshino T, Ohtsu A, Suenaga N, Sato M, Kakizume T, Robson M, Quadt C, Doi T. Phase I dose-escalation study of buparlisib (BKM120), an oral pan-class I PI3K inhibitor, in Japanese patients with advanced solid tumors. Cancer Sci 2014; 105:347-53. [PMID: 24405565 PMCID: PMC4317947 DOI: 10.1111/cas.12350] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2013] [Revised: 12/19/2013] [Accepted: 12/28/2013] [Indexed: 12/27/2022] Open
Abstract
Buparlisib (BKM120) is an oral pan-phosphatidylinositol 3-kinase inhibitor, targeting all four isoforms of class I PI3K (α, β, γ and δ). This open-label Phase I dose-escalation study was conducted to determine the maximum tolerated dose of continuous daily buparlisib in Japanese patients with advanced solid tumors. Secondary objectives included safety and tolerability, pharmacokinetics, antitumor activity and pharmacodynamic marker changes. Fifteen patients were treated at 25 mg/day (n = 3), 50 mg/day (n = 3) and 100 mg/day (n = 9) dose levels. One dose-limiting toxicity of Grade 4 abnormal liver function occurred at 100 mg/day. Considering the safety profile and the maximum tolerated dose in the first-in-man study of buparlisib in non-Japanese patients, further dose escalation was stopped and 100 mg/day was declared the recommended dose. The most common treatment-related adverse events were rash, abnormal hepatic function (including increased transaminase levels), increased blood insulin levels and increased eosinophil count. Hyperglycemia was experienced by two patients, one Grade 1 and one Grade 4, and mood alterations were experienced by three patients, two Grade 1 and one Grade 2. Pharmacokinetic results showed that buparlisib was rapidly absorbed in a dose-proportional manner. Best overall response was stable disease for six patients, including one unconfirmed partial response. In these Japanese patients with advanced solid tumors, buparlisib had a manageable safety profile, with similar pharmacokinetics to non-Japanese patients. The recommended dose of 100 mg/day will be used in future studies of buparlisib in Japanese patients.
Collapse
|
121
|
Peddi PF, Hurvitz SA. PI3K pathway inhibitors for the treatment of brain metastases with a focus on HER2+ breast cancer. J Neurooncol 2014; 117:7-13. [PMID: 24469856 DOI: 10.1007/s11060-014-1369-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Accepted: 01/15/2014] [Indexed: 11/27/2022]
Abstract
The incidence of breast cancer brain metastases has increased in recent years, largely due to improved control of systemic disease with human epidermal growth factor receptor 2 (HER2)-targeted agents and the inability of most of these agents to efficiently cross the blood-blood barrier (BBB) and control central nervous system disease. There is, therefore, an urgent unmet need for treatments to prevent and treat HER2+ breast cancer brain metastases (BCBMs). Aberrant activation of the phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) signaling pathway is frequently observed in many cancers, including primary breast tumors and BCBMs. Agents targeting key components of this pathway have demonstrated antitumor activity in diverse cancers, and may represent a new treatment strategy for BCBMs. In preclinical studies, several inhibitors of PI3K and mTOR have demonstrated an ability to penetrate the BBB and down-regulate PI3K signaling, indicating that these agents may be potential therapies for brain metastatic disease. The PI3K inhibitor buparlisib (BKM120) and the mTOR inhibitor everolimus (RAD001) are currently under evaluation in combination with trastuzumab in patients with HER2+ BCBMs.
Collapse
Affiliation(s)
- Parvin F Peddi
- Division of Hematology Oncology, University of California, Los Angeles, 10945 Le Conte Avenue, PVUB Suite 3360, Los Angeles, CA, 90095, USA
| | | |
Collapse
|
122
|
Vander Broek R, Mohan S, Eytan DF, Chen Z, Van Waes C. The PI3K/Akt/mTOR axis in head and neck cancer: functions, aberrations, cross-talk, and therapies. Oral Dis 2013; 21:815-25. [DOI: 10.1111/odi.12206] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 11/03/2013] [Accepted: 11/03/2013] [Indexed: 12/14/2022]
Affiliation(s)
- R Vander Broek
- Tumor Biology Section; Head and Neck Surgery Branch; National Institute on Deafness and Other Communication Disorders; National Institutes of Health; Bethesda MD USA
- Medical Research Scholars Program; National Institutes of Health; Bethesda MD USA
- School of Dentistry; University of Michigan; Ann Arbor MI USA
| | - S Mohan
- Tumor Biology Section; Head and Neck Surgery Branch; National Institute on Deafness and Other Communication Disorders; National Institutes of Health; Bethesda MD USA
- Medical Research Scholars Program; National Institutes of Health; Bethesda MD USA
| | - DF Eytan
- Tumor Biology Section; Head and Neck Surgery Branch; National Institute on Deafness and Other Communication Disorders; National Institutes of Health; Bethesda MD USA
- Medical Research Scholars Program; National Institutes of Health; Bethesda MD USA
| | - Z Chen
- Tumor Biology Section; Head and Neck Surgery Branch; National Institute on Deafness and Other Communication Disorders; National Institutes of Health; Bethesda MD USA
| | - C Van Waes
- Tumor Biology Section; Head and Neck Surgery Branch; National Institute on Deafness and Other Communication Disorders; National Institutes of Health; Bethesda MD USA
| |
Collapse
|
123
|
Polivka J, Janku F. Molecular targets for cancer therapy in the PI3K/AKT/mTOR pathway. Pharmacol Ther 2013; 142:164-75. [PMID: 24333502 DOI: 10.1016/j.pharmthera.2013.12.004] [Citation(s) in RCA: 595] [Impact Index Per Article: 49.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Accepted: 11/22/2013] [Indexed: 12/20/2022]
Abstract
Aberrations in various cellular signaling pathways are instrumental in regulating cellular metabolism, tumor development, growth, proliferation, metastasis and cytoskeletal reorganization. The fundamental cellular signaling cascade involved in these processes, the phosphatidylinositol 3-kinase/protein kinase-B/mammalian target of rapamycin (PI3K/AKT/mTOR), closely related to the mitogen-activated protein kinase (MAPK) pathway, is a crucial and intensively explored intracellular signaling pathway in tumorigenesis. Various activating mutations in oncogenes together with the inactivation of tumor suppressor genes are found in diverse malignancies across almost all members of the pathway. Substantial progress in uncovering PI3K/AKT/mTOR alterations and their roles in tumorigenesis has enabled the development of novel targeted molecules with potential for developing efficacious anticancer treatment. Two approved anticancer drugs, everolimus and temsirolimus, exemplify targeted inhibition of PI3K/AKT/mTOR in the clinic and many others are in preclinical development as well as being tested in early clinical trials for many different types of cancer. This review focuses on targeted PI3K/AKT/mTOR signaling from the perspective of novel molecular targets for cancer therapy found in key pathway members and their corresponding experimental therapeutic agents. Various aberrant prognostic and predictive biomarkers are also discussed and examples are given. Novel approaches to PI3K/AKT/mTOR pathway inhibition together with a better understanding of prognostic and predictive markers have the potential to significantly improve the future care of cancer patients in the current era of personalized cancer medicine.
Collapse
Affiliation(s)
- Jiri Polivka
- Department of Histology and Embryology and Biomedical Centre, Faculty of Medicine Plzen, Charles University Prague, Husova 3, 301 66 Plzen, Czech Republic; Department of Neurology, Faculty Hospital Plzen, Alej Svobody 80, 304 60 Plzen, Czech Republic
| | - Filip Janku
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA.
| |
Collapse
|
124
|
Abstract
The PI3K pathway is over-activated in the majority of human cancers. This may occur through oncogenic activation of upstream RAS isoforms and tyrosine kinase receptors, or by mutational activation of components of the PI3K pathway themselves. Stimulation of the PI3K pathway enhances growth, survival, and metabolism of cancer cells. Migration, invasion, and angiogenesis are also supported by PI3K signaling. Thus, the PI3K pathway is an attractive candidate for the therapeutic targeting of tumors. Multiple kinases within the PI3Ks, AKT, and mTOR pathway have been selected for inhibition, and dual inhibitors have also been produced. Recently, the development of kinase inhibitors with enhanced specificity and improved pharmacokinetics has facilitated the investigation of PI3K pathway inhibition in clinical trials. Initial reports are encouraging, with tolerable toxicity profiles reported. PI3K inhibitors have provided some benefit as single-agent treatments of advanced solid tumors and the possibilities for enhanced effect with combination treatments look promising. In this chapter, we describe the PI3K inhibitors currently under investigation for the treatment of cancer and discuss the opportunities and obstacles that have been revealed by the latest preclinical and clinical studies.
Collapse
Affiliation(s)
- Clare Sheridan
- Signal Transduction Laboratory, Cancer Research UK London Research Institute, London, United Kingdom.
| | - Julian Downward
- Signal Transduction Laboratory, Cancer Research UK London Research Institute, London, United Kingdom; Lung Cancer Group, Division of Cancer Biology, The Institute of Cancer Research, London, United Kingdom.
| |
Collapse
|
125
|
Matsushima-Nishiwaki R, Kumada T, Nagasawa T, Suzuki M, Yasuda E, Okuda S, Maeda A, Kaneoka Y, Toyoda H, Kozawa O. Direct association of heat shock protein 20 (HSPB6) with phosphoinositide 3-kinase (PI3K) in human hepatocellular carcinoma: regulation of the PI3K activity. PLoS One 2013; 8:e78440. [PMID: 24223153 PMCID: PMC3819392 DOI: 10.1371/journal.pone.0078440] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 09/11/2013] [Indexed: 11/18/2022] Open
Abstract
HSP20 (HSPB6), one of small heat shock proteins (HSPs), is constitutively expressed in various tissues and has several functions. We previously reported that the expression levels of HSP20 in human hepatocellular carcinoma (HCC) cells inversely correlated with the progression of HCC, and that HSP20 suppresses the growth of HCC cells via the AKT and mitogen-activated protein kinase signaling pathways. However, the exact mechanism underlying the effect of HSP20 on the regulation of these signaling pathways remains to be elucidated. To clarify the details of this effect in HCC, we explored the direct targets of HSP20 in HCC using human HCC-derived HuH7 cells with HSP20 overexpression. HSP20 proteins in the HuH7 cells were coimmunoprecipitated with the p85 regulatory subunit and p110 catalytic subunit of phosphoinositide 3-kinase (PI3K), an upstream kinase of AKT. Although HSP20 overexpression in HCC cells failed to affect the expression levels of PI3K, the activity of PI3K in the unstimulated cells and even in the transforming growth factor-α stimulated cells were downregulated by HSP20 overexpression. The association of HSP20 with PI3K was also observed in human HCC tissues in vivo. These findings strongly suggest that HSP20 directly associates with PI3K and suppresses its activity in HCC, resulting in the inhibition of the AKT pathway, and subsequently decreasing the growth of HCC.
Collapse
Affiliation(s)
| | - Takashi Kumada
- Department of Gastroenterology, Ogaki Municipal Hospital, Ogaki, Gifu, Japan
| | - Tomoaki Nagasawa
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Mariko Suzuki
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Eisuke Yasuda
- Department of Radiological Technology, Suzuka University of Medical Science, Suzuka, Mie, Japan
| | - Seiji Okuda
- Department of Medical Technology, Ogaki Municipal Hospital, Ogaki, Gifu, Japan
| | - Atsuyuki Maeda
- Department of Surgery, Ogaki Municipal Hospital, Ogaki, Gifu, Japan
| | - Yuji Kaneoka
- Department of Surgery, Ogaki Municipal Hospital, Ogaki, Gifu, Japan
| | - Hidenori Toyoda
- Department of Gastroenterology, Ogaki Municipal Hospital, Ogaki, Gifu, Japan
| | - Osamu Kozawa
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu, Japan
- * E-mail:
| |
Collapse
|
126
|
Shapiro GI, Rodon J, Bedell C, Kwak EL, Baselga J, Braña I, Pandya SS, Scheffold C, Laird AD, Nguyen LT, Xu Y, Egile C, Edelman G. Phase I safety, pharmacokinetic, and pharmacodynamic study of SAR245408 (XL147), an oral pan-class I PI3K inhibitor, in patients with advanced solid tumors. Clin Cancer Res 2013; 20:233-45. [PMID: 24166903 DOI: 10.1158/1078-0432.ccr-13-1777] [Citation(s) in RCA: 126] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE SAR245408 is a pan-class I phosphoinositide 3-kinase (PI3K) inhibitor. This phase I study determined the maximum tolerated dose (MTD) of two dosing schedules [first 21 days of a 28-day period (21/7) and continuous once-daily dosing (CDD)], pharmacokinetic and pharmacodynamic profiles, and preliminary efficacy. EXPERIMENTAL DESIGN Patients with refractory advanced solid malignancies were treated with SAR245408 using a 3 + 3 design. Pharmacokinetic parameters were determined after single and repeated doses. Pharmacodynamic effects were evaluated in plasma, hair sheath cells, and skin and tumor biopsies. RESULTS Sixty-nine patients were enrolled. The MTD of both schedules was 600 mg; dose-limiting toxicities were maculopapular rash and hypersensitivity reaction. The most frequent drug-related adverse events included dermatologic toxicities, diarrhea, nausea, and decreased appetite. Plasma pharmacokinetics showed a median time to maximum concentration of 8 to 22 hours, mean terminal elimination half-life of 70 to 88 hours, and 5- to 13-fold accumulation after daily dosing (first cycle). Steady-state concentration was reached between days 15 and 21, and exposure was dose-proportional with doses up to 400 mg. SAR245408 inhibited the PI3K pathway (∼40%-80% reduction in phosphorylation of AKT, PRAS40, 4EBP1, and S6 in tumor and surrogate tissues) and, unexpectedly, also inhibited the MEK/ERK pathway. A partial response was seen in one patient with advanced non-small cell lung cancer. Eight patients were progression-free at 6 months. Pharmacodynamic and clinical activity were observed irrespective of tumor PI3K pathway molecular alterations. CONCLUSIONS SAR245408 was tolerable at doses associated with PI3K pathway inhibition. The recommended phase II dose of the capsule formulation is 600 mg administered orally with CDD.
Collapse
Affiliation(s)
- Geoffrey I Shapiro
- Authors' Affiliations: Dana-Farber Cancer Institute; Massachusetts General Hospital; Beth Israel Deaconess Medical Center, Boston, Massachusetts; Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain; Mary Crowley Cancer Research Centers, Dallas, Texas; Exelixis Inc., South San Francisco, California; and Sanofi, Cambridge, Massachusetts, and Vitry-sur-Seine, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
127
|
Bowles DW, Ma WW, Senzer N, Brahmer JR, Adjei AA, Davies M, Lazar AJ, Vo A, Peterson S, Walker L, Hausman D, Rudin CM, Jimeno A. A multicenter phase 1 study of PX-866 in combination with docetaxel in patients with advanced solid tumours. Br J Cancer 2013; 109:1085-92. [PMID: 23942080 PMCID: PMC3778312 DOI: 10.1038/bjc.2013.474] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 06/23/2013] [Accepted: 07/23/2013] [Indexed: 12/31/2022] Open
Abstract
Background: This phase I, dose-finding study determined the safety, maximum tolerated dose (MTD)/recommended phase 2 dose (RP2D), pharmacokinetics, and antitumour activity of PX-866, a phosphatidylinositol 3-kinase inhibitor, combined with docetaxel in patients with incurable solid tumours. Methods: PX-866 was administered at escalating doses (4–8 mg daily) with docetaxel 75 mg m−2 intravenously every 21 days. Archived tumour tissue was assessed for potential predictive biomarkers. Results: Forty-three patients were enrolled. Most adverse events (AEs) were grade 1 or 2. The most frequent study drug-related AE was diarrhoea (76.7%), with gastrointestinal disorders occurring in 79.1% (docetaxel-related) and 83.7% (PX-866-related). No dose-limiting toxicities were observed. The RP2D was 8 mg, the same as the single-agent MTD. Co-administration of PX-866 and docetaxel did not affect either drug's PKs. Best responses in 35 evaluable patients were: 2 partial responses (6%), 22 stable disease (63%), and 11 disease progression (31%). Eleven patients remained on study for >180 days, including 8 who maintained disease control on single-agent PX-866. Overall median progression-free survival (PFS) was 73.5 days (range: 1–569). A non-significant association between longer PFS for PIK3CA-MUT/KRAS-WT vs PIK3CA-WT/KRAS-WT was observed. Conclusion: Treatment with PX-866 and docetaxel was well tolerated, without evidence of overlapping/cumulative toxicity. Further investigation with this combination is justified.
Collapse
Affiliation(s)
- D W Bowles
- Division of Medical Oncology, School of Medicine, Universitiy of Colorado, 12801 E. 17th Avenue, MS 8117, Aurora, CO 80045, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
128
|
Keysar SB, Astling DP, Anderson RT, Vogler BW, Bowles DW, Morton JJ, Paylor JJ, Glogowska MJ, Le PN, Eagles-Soukup JR, Kako SL, Takimoto SM, Sehrt DB, Umpierrez A, Pittman MA, Macfadden SM, Helber RM, Peterson S, Hausman DF, Said S, Leem TH, Goddard JA, Arcaroli JJ, Messersmith WA, Robinson WA, Hirsch FR, Varella-Garcia M, Raben D, Wang XJ, Song JI, Tan AC, Jimeno A. A patient tumor transplant model of squamous cell cancer identifies PI3K inhibitors as candidate therapeutics in defined molecular bins. Mol Oncol 2013; 7:776-90. [PMID: 23607916 PMCID: PMC3760013 DOI: 10.1016/j.molonc.2013.03.004] [Citation(s) in RCA: 126] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Revised: 03/23/2013] [Accepted: 03/25/2013] [Indexed: 12/23/2022] Open
Abstract
Targeted therapy development in head and neck squamous cell carcinoma (HNSCC) is challenging given the rarity of activating mutations. Additionally, HNSCC incidence is increasing related to human papillomavirus (HPV). We sought to develop an in vivo model derived from patients reflecting the evolving HNSCC epidemiologic landscape, and use it to identify new therapies. Primary and relapsed tumors from HNSCC patients, both HPV+ and HPV-, were implanted on mice, giving rise to 25 strains. Resulting xenografts were characterized by detecting key mutations, measuring protein expression by IHC and gene expression/pathway analysis by mRNA-sequencing. Drug efficacy studies were run with representative xenografts using the approved drug cetuximab as well as the new PI3K inhibitor PX-866. Tumors maintained their original morphology, genetic profiles and drug susceptibilities through serial passaging. The genetic makeup of these tumors was consistent with known frequencies of TP53, PI3KCA, NOTCH1 and NOTCH2 mutations. Because the EGFR inhibitor cetuximab is a standard HNSCC therapy, we tested its efficacy and observed a wide spectrum of efficacy. Cetuximab-resistant strains had higher PI3K/Akt pathway gene expression and protein activation than cetuximab-sensitive strains. The PI3K inhibitor PX-866 had anti-tumor efficacy in HNSCC models with PIK3CA alterations. Finally, PI3K inhibition was effective in two cases with NOTCH1 inactivating mutations. In summary, we have developed an HNSCC model covering its clinical spectrum whose major genetic alterations and susceptibility to anticancer agents represent contemporary HNSCC. This model enables to prospectively test therapeutic-oriented hypotheses leading to personalized medicine.
Collapse
Affiliation(s)
- Stephen B. Keysar
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine (UCSOM), Aurora, CO 80045, United States
| | - David P. Astling
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine (UCSOM), Aurora, CO 80045, United States
- Department of Biostatistics and Informatics, UCSOM, CO 80045, United States
| | - Ryan T. Anderson
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine (UCSOM), Aurora, CO 80045, United States
| | - Brian W. Vogler
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine (UCSOM), Aurora, CO 80045, United States
| | - Daniel W. Bowles
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine (UCSOM), Aurora, CO 80045, United States
| | - J. Jason Morton
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine (UCSOM), Aurora, CO 80045, United States
| | - Jeramiah J. Paylor
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine (UCSOM), Aurora, CO 80045, United States
| | - Magdalena J. Glogowska
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine (UCSOM), Aurora, CO 80045, United States
| | - Phuong N. Le
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine (UCSOM), Aurora, CO 80045, United States
| | - Justin R. Eagles-Soukup
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine (UCSOM), Aurora, CO 80045, United States
| | - Severine L. Kako
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine (UCSOM), Aurora, CO 80045, United States
| | - Sarah M. Takimoto
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine (UCSOM), Aurora, CO 80045, United States
| | - Daniel B. Sehrt
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine (UCSOM), Aurora, CO 80045, United States
| | - Adrian Umpierrez
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine (UCSOM), Aurora, CO 80045, United States
| | - Morgan A. Pittman
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine (UCSOM), Aurora, CO 80045, United States
| | - Sarah M. Macfadden
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine (UCSOM), Aurora, CO 80045, United States
| | - Ryan M. Helber
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine (UCSOM), Aurora, CO 80045, United States
| | | | | | - Sherif Said
- Department of Pathology, UCSOM, CO 80045, United States
| | - Ted H. Leem
- Department of Otolaryngology, UCSOM, CO 80045, United States
| | | | - John J. Arcaroli
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine (UCSOM), Aurora, CO 80045, United States
| | - Wells A. Messersmith
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine (UCSOM), Aurora, CO 80045, United States
| | - William A. Robinson
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine (UCSOM), Aurora, CO 80045, United States
| | - Fred R. Hirsch
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine (UCSOM), Aurora, CO 80045, United States
| | - Marileila Varella-Garcia
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine (UCSOM), Aurora, CO 80045, United States
| | - David Raben
- Department of Radiation Oncology, USCOM, CO 80045, United States
| | - Xiao-Jing Wang
- Department of Pathology, UCSOM, CO 80045, United States
- Charles C. Gates Center for Stem Cell Biology, UCSOM, CO 80045, United States
| | - John I. Song
- Department of Otolaryngology, UCSOM, CO 80045, United States
| | - Aik-Choon Tan
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine (UCSOM), Aurora, CO 80045, United States
- Department of Biostatistics and Informatics, UCSOM, CO 80045, United States
| | - Antonio Jimeno
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine (UCSOM), Aurora, CO 80045, United States
- Department of Otolaryngology, UCSOM, CO 80045, United States
- Charles C. Gates Center for Stem Cell Biology, UCSOM, CO 80045, United States
| |
Collapse
|
129
|
Sajithlal GB, Hamed HA, Cruickshanks N, Booth L, Tavallai S, Syed J, Grant S, Poklepovic A, Dent P. Sorafenib/regorafenib and phosphatidyl inositol 3 kinase/thymoma viral proto-oncogene inhibition interact to kill tumor cells. Mol Pharmacol 2013; 84:562-71. [PMID: 23877009 DOI: 10.1124/mol.113.088005] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The present studies were undertaken to determine whether the multikinase inhibitors sorafenib/regorafenib cooperated with clinically relevant , phosphatidyl inositol 3 kinase (PI3K)-thymoma viral proto-oncogene (AKT) inhibitors to kill tumor cells. In liver, colorectal, lung, breast, kidney, and brain cancer cells, at clinically achievable doses, sorafenib/regorafenib and the PI3K inhibitor acetic acid (1S,4E,10R,11R,13S,14R)-[4-diallylaminomethylene-6-hydroxy-1-methoxymethyl-10,13-dimethyl-3,7,17-trioxo-1,3,4,7,10,11,12,13,14,15,16,17-dodecahydro-2-oxa-cyclopenta[a]phenanthren-11-yl ester (PX-866) cooperated in a greater than additive fashion to kill tumor cells. Cells lacking phosphatase and tensin homolog were as sensitive to the drug combination as cells expressing the protein. Similar data were obtained using the AKT inhibitors perifosine and 8-[4-(1-aminocyclobutyl)phenyl]-9-phenyl-1,2,4-triazolo[3,4-f] [1,6]naphthyridin-3(2H)-one hydrochloride (MK2206). PX-866 treatment abolished AKT/glycogen synthase kinase 3 (GSK3) phosphorylation, and cell killing correlated with reduced activity of AKT and mammalian target of rapamycin (mTOR). Expression of activated AKT and to a lesser extent activated mTOR reduced drug combination lethality. Expression of B-cell lymphoma-extra large or dominant negative caspase 9, but not cellular FLICE (FADD-like IL-1b-converting enzyme)-inhibitory protein short, protected cells from the drug combination. Treatment of cells with PX-866 increased protein levels of p62, lysosome-associated membrane protein 2 (LAMP2), and microtubule-associated protein light chain (LC) 3 and LC3II that correlated with a large increase in LC3-green fluorescent protein (GFP) vesicle numbers. Exposure of PX-866 treated cells to sorafenib reduced p62 and LAMP2 levels, decreased the ratio of LC3 to LC3II, and reduced LC3-GFP vesicle levels. Knockdown of Beclin1 or autophagy-related 5 suppressed drug toxicity by ∼40%. In vivo, sorafenib and PX-866 or regorafenib and MK2206 cooperated to suppress the growth of established HuH7 and HCT116 tumors, respectively. Collectively our data demonstrate that the combination of sorafenib family kinase inhibitors with inhibitors of the PI3K/AKT pathway kills tumor cells in vitro and in vivo.
Collapse
Affiliation(s)
- Gangadharan B Sajithlal
- Departments of Neurosurgery (G.B.S., H.A.H., N.C., L.B., S.T., J.S., P.D.) and Medicine (S.G., A.P.), Virginia Commonwealth University, Richmond, Virginia
| | | | | | | | | | | | | | | | | |
Collapse
|
130
|
Abstract
INTRODUCTION Colorectal cancer (CRC) is a leading cause of cancer mortality worldwide, with > 1.2 million new cases and > 600,000 deaths per year. This complex disease is driven by multiple genetic lesions, commonly dysregulated signaling pathways, and aberrant activity of developmental programs such as Notch and Wnt. While emerging therapies such as EGFR inhibitors are improving treatment regimens, recent findings elucidating the role of cancer stem cells provide insights into opportunities for novel therapeutic intervention. AREAS COVERED This review provides a background on CRC statistics, colon anatomy and CRC pathobiology, CRC genetics and current and emerging therapies. Furthermore, the article discusses the role of developmental signaling pathways governing self-renewal biology as potential points for therapeutic intervention. EXPERT OPINION Despite recent advances including the introduction of targeted therapeutics, prognosis for advanced CRC patients remains bleak, reinforcing the need for novel therapeutic intervention. Developmental pathways such as Notch and Wnt provide opportunities to address this urgent need, and preclinical evidence supports targeting these pathways in CRC. Progress has been made toward this end, and while challenges persist, an increasing number of preclinical findings show promise.
Collapse
Affiliation(s)
- Joshua C Curtin
- Oncology Drug Discovery, Research and Development, Bristol-Myers Squibb, Route 206 and Provinceline Road, Princeton, NJ, USA.
| |
Collapse
|
131
|
BRAF, KRAS, and Phosphatidylinositol 3-Kinase in the Management of Metastatic Colorectal Cancer. CURRENT COLORECTAL CANCER REPORTS 2013. [DOI: 10.1007/s11888-012-0152-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
132
|
Brana I, Siu LL. Clinical development of phosphatidylinositol 3-kinase inhibitors for cancer treatment. BMC Med 2012; 10:161. [PMID: 23232172 PMCID: PMC3552942 DOI: 10.1186/1741-7015-10-161] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Accepted: 12/11/2012] [Indexed: 01/21/2023] Open
Abstract
The phosphatidylinositol 3-kinase (PI3K) pathway is commonly deregulated in cancer. In recent years, the results of the first phase I clinical trials with PI3K inhibitors have become available. In comparison to other targeted agents such v-raf murine sarcoma viral oncogene homolog B1 (BRAF) inhibitors in melanoma or crizotinib in anaplastic lymphoma receptor tyrosine kinase (ALK) translocated tumors, the number of objective responses to PI3K inhibitors is less dramatic. In this review we propose possible strategies to optimize the clinical development of PI3K inhibitors: by exploring the potential role of PI3K isoform-specific inhibitors in improving the therapeutic index, molecular characterization as a basis for patient selection, and the relevance of performing serial tumor biopsies to understand the associated mechanisms of drug resistance. The main focus of this review will be on PI3K isoform-specific inhibitors by describing the functions of different PI3K isoforms, the preclinical activity of selective PI3K isoform-specific inhibitors and the early clinical data of these compounds.
Collapse
Affiliation(s)
- Irene Brana
- Drug Development Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada
| | - Lillian L Siu
- Drug Development Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada
| |
Collapse
|
133
|
Nardi V, Sadow PM, Juric D, Zhao D, Cosper AK, Bergethon K, Scialabba VL, Batten JM, Borger DR, Iafrate AJ, Heist RS, Lawrence DP, Flaherty KT, Bendell JC, Deschler D, Li Y, Wirth LJ, Dias-Santagata D. Detection of novel actionable genetic changes in salivary duct carcinoma helps direct patient treatment. Clin Cancer Res 2012. [PMID: 23186780 DOI: 10.1158/1078-0432.ccr-12-1842] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Salivary duct carcinomas (SDC) are a rare and aggressive subtype of salivary gland cancers for which cytotoxic chemotherapy has limited efficacy. We investigated whether genotyping analysis could detect novel tumor-specific mutations that would help direct SDC patient treatment using targeted agents. EXPERIMENTAL DESIGN We genotyped 27 SDC archival specimens from patients followed at Massachusetts General Hospital and Massachusetts Eye and Ear Infirmary (Boston, MA) between 2000 and 2011. These included the tumors of 8 patients who were tested prospectively. Targeted mutational analysis of 13 clinically relevant cancer genes was conducted using SNaPshot multiplexed genotyping. FISH was conducted to detect HER2 gene amplification. Patient medical records and tumor histopathologic features were retrospectively reviewed. RESULTS Mutually exclusive genetic aberrations were detected in 15 of 27 (56%) tumors, including 2 (7%) mutations in BRAF, 5 (19%) mutations in PIK3CA, and 8 (30%) cases of HER2 gene amplification. To our knowledge, this is the first time that BRAF and PIK3CA mutations have been reported in this tumor type. Prospective clinical testing of 8 patients with SDC identified actionable genetic alterations in 6 tumors and influenced therapeutic decisions for all 6 patients. CONCLUSION SNaPshot molecular profiling identified novel genetic changes in SDCs, expanded the therapeutic options for patients with this rare tumor, and is changing SDC management at our institution. These findings highlight the importance of using broad-based genetic profiling to expedite the identification of effective-targeted therapies for patients with rare malignancies.
Collapse
Affiliation(s)
- Valentina Nardi
- Departments of Pathology and Medicine, Massachusetts General Hospital; Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
134
|
Jeong AJ, Chung CN, Kim HJ, Bae KS, Choi S, Jun WJ, Shim SI, Kang TH, Leem SH, Chung JW. Gecko Proteins Exert Anti-Tumor Effect against Cervical Cancer Cells Via PI3-Kinase/Akt Pathway. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2012; 16:361-5. [PMID: 23118562 PMCID: PMC3484523 DOI: 10.4196/kjpp.2012.16.5.361] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Accepted: 09/17/2012] [Indexed: 12/13/2022]
Abstract
Anti-tumor activity of the proteins from Gecko (GP) on cervical cancer cells, and its signaling mechanisms were assessed by viable cell counting, propidium iodide (PI) staining, and Western blot analysis. GP induced the cell death of HeLa cells in a dose-dependent manner while it did not affect the viability of normal cells. Western blot analysis showed that GP decreased the activation of Akt, and co-administration of GP and Akt inhibitors synergistically exerted anti-tumor activities on HeLa cells, suggesting the involvement of PI3-kinase/Akt pathway in GP-induced cell death of the cancer cells. Indeed, the cytotoxic effect of GP against HeLa cells was inhibited by overexpression of constituvely active form of Akt in HeLa cells. The candidates of the functional proteins in GP were analyzed by Mass-spectrum. Taken together, our results suggest that GP elicits anti-tumor activity against HeLa cells by inhibition of PI3-kinase/Akt pathway.
Collapse
Affiliation(s)
- Ae-Jin Jeong
- Department of Biological Science, Dong-A University, Busan 604-714, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|