1
|
Castro MP, Khanlou N, Fallah A, Pampana A, Alam A, Lala DA, Roy KGG, Amara ARR, Prakash A, Singh D, Behura L, Kumar A, Kapoor S. Targeting chromosome 12q amplification in relapsed glioblastoma: the use of computational biological modeling to identify effective therapy-a case report. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1289. [PMID: 36618786 PMCID: PMC9816820 DOI: 10.21037/atm-2022-62] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 10/20/2022] [Indexed: 11/17/2022]
Abstract
Background Relapsed glioblastoma (GBM) is often an imminently fatal condition with limited therapeutic options. Computation biological modeling, i.e., biosimulation, of comprehensive genomic information affords the opportunity to create a disease avatar that can be interrogated in silico with various drug combinations to identify the most effective therapies. Case Description We report the outcome of a GBM patient with chromosome 12q amplification who achieved substantial disease remission from a novel therapy using this approach. Following next generation sequencing (NGS) was performed on the tumor specimen. Mutation and copy number changes were input into a computational biologic model to create an avatar of disease behavior and the malignant phenotype. In silico responses to various drug combinations were biosimulated in the disease network. Efficacy scores representing the computational effect of treatment for each strategy were generated and compared to each other to ascertain the differential benefit in drug response from various regimens. Biosimulation identified CDK4/6 inhibitors, nelfinavir and leflunomide to be effective agents singly and in combination. Upon receiving this treatment, the patient achieved a prompt and clinically meaningful remission lasting 6 months. Conclusions Biosimulation has utility to identify active treatment combinations, stratify treatment options and identify investigational agents relevant to patients' comprehensive genomic abnormalities. Additionally, the combination of abemaciclib and nelfinavir appear promising for GBM and potentially other cancers harboring chromosome 12q amplification.
Collapse
Affiliation(s)
- Michael P. Castro
- Beverly Hills Cancer Center and Personalized Cancer Medicine, PLLC, Beverly Hills, CA, USA;,Cellworks Group, Inc., S. San Francisco, CA, USA;,Cellworks Group, Inc., Bangalore, India
| | - Negar Khanlou
- Department of Pathology, Ronald Reagan UCLA Medical Center, Los Angeles, CA, USA
| | - Aria Fallah
- Department of Neurosurgery, Ronald Reagan UCLA Medical Center, Los Angeles, CA, USA
| | - Anusha Pampana
- Cellworks Group, Inc., S. San Francisco, CA, USA;,Cellworks Group, Inc., Bangalore, India
| | - Aftab Alam
- Cellworks Group, Inc., S. San Francisco, CA, USA;,Cellworks Group, Inc., Bangalore, India
| | - Deepak Anil Lala
- Cellworks Group, Inc., S. San Francisco, CA, USA;,Cellworks Group, Inc., Bangalore, India
| | - Kunal Ghosh Ghosh Roy
- Cellworks Group, Inc., S. San Francisco, CA, USA;,Cellworks Group, Inc., Bangalore, India
| | - Anish Raju R. Amara
- Cellworks Group, Inc., S. San Francisco, CA, USA;,Cellworks Group, Inc., Bangalore, India
| | - Annapoorna Prakash
- Cellworks Group, Inc., S. San Francisco, CA, USA;,Cellworks Group, Inc., Bangalore, India
| | - Divya Singh
- Cellworks Group, Inc., S. San Francisco, CA, USA;,Cellworks Group, Inc., Bangalore, India
| | - Liptimayee Behura
- Cellworks Group, Inc., S. San Francisco, CA, USA;,Cellworks Group, Inc., Bangalore, India
| | - Ansu Kumar
- Cellworks Group, Inc., S. San Francisco, CA, USA;,Cellworks Group, Inc., Bangalore, India
| | - Shweta Kapoor
- Cellworks Group, Inc., S. San Francisco, CA, USA;,Cellworks Group, Inc., Bangalore, India
| |
Collapse
|
2
|
Padda SK, Aredo JV, Vali S, Singh NK, Vasista SM, Kumar A, Neal JW, Abbasi T, Wakelee HA. Computational Biological Modeling Identifies PD-(L)1 Immunotherapy Sensitivity Among Molecular Subgroups of KRAS-Mutated Non-Small-Cell Lung Cancer. JCO Precis Oncol 2021; 5:153-162. [PMID: 34994595 DOI: 10.1200/po.20.00172] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
PURPOSE KRAS-mutated (KRASMUT) non-small-cell lung cancer (NSCLC) is emerging as a heterogeneous disease defined by comutations, which may confer differential benefit to PD-(L)1 immunotherapy. In this study, we leveraged computational biological modeling (CBM) of tumor genomic data to identify PD-(L)1 immunotherapy sensitivity among KRASMUT NSCLC molecular subgroups. MATERIALS AND METHODS In this multicohort retrospective analysis, the genotype clustering frequency ranked method was used for molecular clustering of tumor genomic data from 776 patients with KRASMUT NSCLC. These genomic data were input into the CBM, in which customized protein networks were characterized for each tumor. The CBM evaluated sensitivity to PD-(L)1 immunotherapy using three metrics: programmed death-ligand 1 expression, dendritic cell infiltration index (nine chemokine markers), and immunosuppressive biomarker expression index (14 markers). RESULTS Genotype clustering identified eight molecular subgroups and the CBM characterized their shared cancer pathway characteristics: KRASMUT/TP53MUT, KRASMUT/CDKN2A/B/CMUT, KRASMUT/STK11MUT, KRASMUT/KEAP1MUT, KRASMUT/STK11MUT/KEAP1MUT, KRASMUT/PIK3CAMUT, KRAS MUT/ATMMUT, and KRASMUT without comutation. CBM identified PD-(L)1 immunotherapy sensitivity in the KRASMUT/TP53MUT, KRASMUT/PIK3CAMUT, and KRASMUT alone subgroups and resistance in the KEAP1MUT containing subgroups. There was insufficient genomic information to elucidate PD-(L)1 immunotherapy sensitivity by the CBM in the KRASMUT/CDKN2A/B/CMUT, KRASMUT/STK11MUT, and KRASMUT/ATMMUT subgroups. In an exploratory clinical cohort of 34 patients with advanced KRASMUT NSCLC treated with PD-(L)1 immunotherapy, the CBM-assessed overall survival correlated well with actual overall survival (r = 0.80, P < .001). CONCLUSION CBM identified distinct PD-(L)1 immunotherapy sensitivity among molecular subgroups of KRASMUT NSCLC, in line with previous literature. These data provide proof-of-concept that computational modeling of tumor genomics could be used to expand on hypotheses from clinical observations of patients receiving PD-(L)1 immunotherapy and suggest mechanisms that underlie PD-(L)1 immunotherapy sensitivity.
Collapse
Affiliation(s)
- Sukhmani K Padda
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | - Jacqueline V Aredo
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | | | | | | | - Ansu Kumar
- Cellworks Research India Pvt Ltd, Bangalore, India
| | - Joel W Neal
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | | | - Heather A Wakelee
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
3
|
Hellyer JA, Ouseph MM, Padda SK, Wakelee HA. Everolimus in the treatment of metastatic thymic epithelial tumors. Lung Cancer 2020; 149:97-102. [PMID: 33007678 DOI: 10.1016/j.lungcan.2020.09.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/05/2020] [Accepted: 09/09/2020] [Indexed: 02/07/2023]
Abstract
INTRODUCTION There is emerging evidence to support the use of mTOR inhibitor everolimus in patients with advanced, relapsed-refractory thymic epithelial tumors (TETs). However, patient selection and identifying predictive biomarkers of response remains a challenge. Here, we describe a single-center experience with everolimus in patients with TETs and provide detailed molecular analysis of their thymic tumors. MATERIALS AND METHODS Data on all patients with advanced TETs who were prescribed everolimus at Stanford University were retrospectively assessed. Time to treatment failure (TTF) and overall survival (OS) were calculated. STAMP, a 130-gene targeted next generation sequencing (NGS) panel, was performed on each tumor sample. RESULTS Twelve patients with thymoma (T) and three with thymic carcinoma (TC) treated with everolimus were included. Patients had been heavily pre-treated with an average of three prior lines of therapy. Three patients discontinued treatment due to adverse events. The average TTF was 14.7 months in T and 2.6 months in TC with median OS of 27.6 months in the entire cohort (NR T and 5.3 months TC). Two patients with paraneoplastic autoimmune diseases had improvement in autoimmunity on everolimus. Pathogenic mutations were observed in 4/15 (27 %) of patients and includedTP53, KEAP1 and CDKN2A. Several variants of unknown significance in key genes responsible for modulating tumor response to mTOR inhibition were also found. CONCLUSION As previously reported in a prospective trial, patients with previously treated advanced TETs appear to benefit from everolimus in this single institution cohort. Moreover, there was a manageable toxicity profile and no cases of everolimus-induced pneumonitis. A targeted NGS panel revealed several pathogenic mutations but there was no association between detectable tumor mutations and time to treatment failure in this cohort.
Collapse
Affiliation(s)
- Jessica A Hellyer
- Stanford Cancer Institute/Stanford University School of Medicine, Stanford, CA, USA
| | - Madhu M Ouseph
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Sukhmani K Padda
- Stanford Cancer Institute/Stanford University School of Medicine, Stanford, CA, USA
| | - Heather A Wakelee
- Stanford Cancer Institute/Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
4
|
A genomics-informed computational biology platform prospectively predicts treatment responses in AML and MDS patients. Blood Adv 2020; 3:1837-1847. [PMID: 31208955 DOI: 10.1182/bloodadvances.2018028316] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 03/12/2019] [Indexed: 12/14/2022] Open
Abstract
Patients with myelodysplastic syndromes (MDS) or acute myeloid leukemia (AML) are generally older and have more comorbidities. Therefore, identifying personalized treatment options for each patient early and accurately is essential. To address this, we developed a computational biology modeling (CBM) and digital drug simulation platform that relies on somatic gene mutations and gene CNVs found in malignant cells of individual patients. Drug treatment simulations based on unique patient-specific disease networks were used to generate treatment predictions. To evaluate the accuracy of the genomics-informed computational platform, we conducted a pilot prospective clinical study (NCT02435550) enrolling confirmed MDS and AML patients. Blinded to the empirically prescribed treatment regimen for each patient, genomic data from 50 evaluable patients were analyzed by CBM to predict patient-specific treatment responses. CBM accurately predicted treatment responses in 55 of 61 (90%) simulations, with 33 of 61 true positives, 22 of 61 true negatives, 3 of 61 false positives, and 3 of 61 false negatives, resulting in a sensitivity of 94%, a specificity of 88%, and an accuracy of 90%. Laboratory validation further confirmed the accuracy of CBM-predicted activated protein networks in 17 of 19 (89%) samples from 11 patients. Somatic mutations in the TET2, IDH1/2, ASXL1, and EZH2 genes were discovered to be highly informative of MDS response to hypomethylating agents. In sum, analyses of patient cancer genomics using the CBM platform can be used to predict precision treatment responses in MDS and AML patients.
Collapse
|
5
|
Rahman R, Trippa L, Alden S, Fell G, Abbasi T, Mundkur Y, Singh NK, Talawdekar A, Husain Z, Vali S, Ligon KL, Wen PY, Alexander BM. Prediction of Outcomes with a Computational Biology Model in Newly Diagnosed Glioblastoma Patients Treated with Radiation Therapy and Temozolomide. Int J Radiat Oncol Biol Phys 2020; 108:716-724. [PMID: 32417407 DOI: 10.1016/j.ijrobp.2020.05.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 04/16/2020] [Accepted: 05/07/2020] [Indexed: 01/18/2023]
Abstract
PURPOSE Precision medicine has been most successful in targeting single mutations, but personalized medicine using broader genomic tumor profiles for individual patients is less well developed. We evaluate a genomics-informed computational biology model (CBM) to predict outcomes from standard treatments and to suggest novel therapy recommendations in glioblastoma (GBM). METHODS AND MATERIALS In this retrospective study, 98 patients with newly diagnosed GBM undergoing surgery followed by radiation therapy and temozolomide at a single institution with available genomic data were identified. Incorporating mutational and copy number aberration data, a CBM was used to simulate the response of GBM tumor cells and generate efficacy predictions for radiation therapy (RTeff) and temozolomide (TMZeff). RTeff and TMZeff were evaluated for association with overall survival and progression-free survival in a Cox regression model. To demonstrate a CBM-based individualized therapy strategy, treatment recommendations were generated for each patient by testing a panel of 45 central nervous system-penetrant US Food and Drug Administration-approved agents. RESULTS High RTeff scores were associated with longer survival on univariable analysis (P < .001), which persisted after controlling for age, extent of resection, performance status, MGMT, and IDH status (P = .017). High RTeff patients had a longer overall survival compared with low RTeff patients (median, 27.7 vs 14.6 months). High TMZeff was also associated with longer survival on univariable analysis (P = .007) but did not hold on multivariable analysis, suggesting an interplay with MGMT status. Among predictions of the 3 most efficacious combination therapies for each patient, only 2.4% (7 of 294) of 2-drug recommendations produced by the CBM included TMZ. CONCLUSIONS CBM-based predictions of RT and TMZ effectiveness were associated with survival in patients with newly diagnosed GBM treated with those therapies, suggesting a possible predictive utility. Furthermore, the model was able to suggest novel individualized monotherapies and combinations. Prospective evaluation of such a personalized treatment strategy in clinical trials is needed.
Collapse
Affiliation(s)
- Rifaquat Rahman
- Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Harvard Medical School, Boston, MA
| | - Lorenzo Trippa
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Harvard School of Public Health, Boston, MA
| | | | - Geoffrey Fell
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Harvard School of Public Health, Boston, MA
| | | | | | | | | | - Zakir Husain
- Cellworks Research India Pvt Ltd, Bengaluru, India
| | | | - Keith L Ligon
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Brian M Alexander
- Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Harvard Medical School, Boston, MA.
| |
Collapse
|
6
|
Olgen S, Kotra LP. Drug Repurposing in the Development of Anticancer Agents. Curr Med Chem 2019; 26:5410-5427. [PMID: 30009698 DOI: 10.2174/0929867325666180713155702] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 06/14/2018] [Accepted: 06/28/2018] [Indexed: 01/07/2023]
Abstract
BACKGROUND Research into repositioning known drugs to treat cancer other than the originally intended disease continues to grow and develop, encouraged in part, by several recent success stories. Many of the studies in this article are geared towards repurposing generic drugs because additional clinical trials are relatively easy to perform and the drug safety profiles have previously been established. OBJECTIVE This review provides an overview of anticancer drug development strategies which is one of the important areas of drug restructuring. METHODS Repurposed drugs for cancer treatments are classified by their pharmacological effects. The successes and failures of important repurposed drugs as anticancer agents are evaluated in this review. RESULTS AND CONCLUSION Drugs could have many off-target effects, and can be intelligently repurposed if the off-target effects can be employed for therapeutic purposes. In cancer, due to the heterogeneity of the disease, often targets are quite diverse, hence a number of already known drugs that interfere with these targets could be deployed or repurposed with appropriate research and development.
Collapse
Affiliation(s)
- Sureyya Olgen
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Biruni University, Istanbul, Turkey
| | - Lakshmi P Kotra
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, M5S 3M2, Canada.,Center for Molecular Design and Preformulations, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, M5G 1L7 Canada.,Multi-Organ Transplant Program, Toronto General Hospital, Toronto, Ontario, M5G 1L7 Canada
| |
Collapse
|
7
|
Fischer CL, Bates AM, Lanzel EA, Guthmiller JM, Johnson GK, Singh NK, Kumar A, Vidva R, Abbasi T, Vali S, Xie XJ, Zeng E, Brogden KA. Computational Models Accurately Predict Multi-Cell Biomarker Profiles in Inflammation and Cancer. Sci Rep 2019; 9:10877. [PMID: 31350446 PMCID: PMC6659691 DOI: 10.1038/s41598-019-47381-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 07/15/2019] [Indexed: 01/28/2023] Open
Abstract
Individual computational models of single myeloid, lymphoid, epithelial, and cancer cells were created and combined into multi-cell computational models and used to predict the collective chemokine, cytokine, and cellular biomarker profiles often seen in inflamed or cancerous tissues. Predicted chemokine and cytokine output profiles from multi-cell computational models of gingival epithelial keratinocytes (GE KER), dendritic cells (DC), and helper T lymphocytes (HTL) exposed to lipopolysaccharide (LPS) or synthetic triacylated lipopeptide (Pam3CSK4) as well as multi-cell computational models of multiple myeloma (MM) and DC were validated using the observed chemokine and cytokine responses from the same cell type combinations grown in laboratory multi-cell cultures with accuracy. Predicted and observed chemokine and cytokine responses of GE KER + DC + HTL exposed to LPS and Pam3CSK4 matched 75% (15/20, p = 0.02069) and 80% (16/20, P = 0.005909), respectively. Multi-cell computational models became ‘personalized’ when cell line-specific genomic data were included into simulations, again validated with the same cell lines grown in laboratory multi-cell cultures. Here, predicted and observed chemokine and cytokine responses of MM cells lines MM.1S and U266B1 matched 75% (3/4) and MM.1S and U266B1 inhibition of DC marker expression in co-culture matched 100% (6/6). Multi-cell computational models have the potential to identify approaches altering the predicted disease-associated output profiles, particularly as high throughput screening tools for anti-inflammatory or immuno-oncology treatments of inflamed multi-cellular tissues and the tumor microenvironment.
Collapse
Affiliation(s)
- Carol L Fischer
- Department of Biology, Waldorf University, Forest City, IA, 50436, USA
| | - Amber M Bates
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Emily A Lanzel
- Department of Oral Pathology, Radiology and Medicine, College of Dentistry, University of Iowa, Iowa City, IA, 52242, USA
| | - Janet M Guthmiller
- College of Dentistry, University of Nebraska Medical Center, Lincoln, NE, 68583, USA
| | - Georgia K Johnson
- Department of Periodontics, College of Dentistry, University of Iowa, Iowa City, IA, 52242, USA
| | - Neeraj Kumar Singh
- Cellworks Group Inc., San Jose, CA, 95110, USA.,Cellworks Research India Pvt. Ltd (Wholly owned subsidiary of Cellworks Group Inc.), Bangalore, India
| | - Ansu Kumar
- Cellworks Group Inc., San Jose, CA, 95110, USA.,Cellworks Research India Pvt. Ltd (Wholly owned subsidiary of Cellworks Group Inc.), Bangalore, India
| | - Robinson Vidva
- Cellworks Group Inc., San Jose, CA, 95110, USA.,Cellworks Research India Pvt. Ltd (Wholly owned subsidiary of Cellworks Group Inc.), Bangalore, India
| | - Taher Abbasi
- Cellworks Group Inc., San Jose, CA, 95110, USA.,Cellworks Research India Pvt. Ltd (Wholly owned subsidiary of Cellworks Group Inc.), Bangalore, India
| | - Shireen Vali
- Cellworks Group Inc., San Jose, CA, 95110, USA.,Cellworks Research India Pvt. Ltd (Wholly owned subsidiary of Cellworks Group Inc.), Bangalore, India
| | - Xian Jin Xie
- Division of Biostatistics and Computational Biology, College of Dentistry, University of Iowa, Iowa City, IA, 52242, USA
| | - Erliang Zeng
- Division of Biostatistics and Computational Biology, College of Dentistry, University of Iowa, Iowa City, IA, 52242, USA
| | - Kim A Brogden
- Department of Periodontics, College of Dentistry, University of Iowa, Iowa City, IA, 52242, USA. .,Iowa Institute for Oral Health Research, College of Dentistry, University of Iowa, Iowa City, IA, 52242, USA.
| |
Collapse
|
8
|
Gal J, Milano G, Ferrero JM, Saâda-Bouzid E, Viotti J, Chabaud S, Gougis P, Le Tourneau C, Schiappa R, Paquet A, Chamorey E. Optimizing drug development in oncology by clinical trial simulation: Why and how? Brief Bioinform 2019; 19:1203-1217. [PMID: 28575140 DOI: 10.1093/bib/bbx055] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Indexed: 12/11/2022] Open
Abstract
In therapeutic research, the safety and efficacy of pharmaceutical products are necessarily tested on humans via clinical trials after an extensive and expensive preclinical development period. Methodologies such as computer modeling and clinical trial simulation (CTS) might represent a valuable option to reduce animal and human assays. The relevance of these methods is well recognized in pharmacokinetics and pharmacodynamics from the preclinical phase to postmarketing. However, they are barely used and are poorly regarded for drug approval, despite Food and Drug Administration and European Medicines Agency recommendations. The generalization of CTS could be greatly facilitated by the availability of software for modeling biological systems, by clinical trial studies and hospital databases. Data sharing and data merging raise legal, policy and technical issues that will need to be addressed. Development of future molecules will have to use CTS for faster development and thus enable better patient management. Drug activity modeling coupled with disease modeling, optimal use of medical data and increased computing speed should allow this leap forward. The realization of CTS requires not only bioinformatics tools to allow interconnection and global integration of all clinical data but also a universal legal framework to protect the privacy of every patient. While recognizing that CTS can never replace 'real-life' trials, they should be implemented in future drug development schemes to provide quantitative support for decision-making. This in silico medicine opens the way to the P4 medicine: predictive, preventive, personalized and participatory.
Collapse
Affiliation(s)
- Jocelyn Gal
- Epidemiology and Biostatistics Unit at the Antoine Lacassagne Center, Nice, France
| | | | | | | | | | | | - Paul Gougis
- Pitie´-Salp^etrie`re Hospital in Paris, France
| | | | | | - Agnes Paquet
- Molecular and Cellular Pharmacology Institute of Sophia Antipolis, Valbonne, France
| | | |
Collapse
|
9
|
Vizirianakis IS, Miliotou AN, Mystridis GA, Andriotis EG, Andreadis II, Papadopoulou LC, Fatouros DG. Tackling pharmacological response heterogeneity by PBPK modeling to advance precision medicine productivity of nanotechnology and genomics therapeutics. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2019. [DOI: 10.1080/23808993.2019.1605828] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Ioannis S. Vizirianakis
- Laboratory of Pharmacology, School of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Androulla N. Miliotou
- Laboratory of Pharmacology, School of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - George A. Mystridis
- Laboratory of Pharmacology, School of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Eleftherios G. Andriotis
- Laboratory of Pharmaceutical Technology, School of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ioannis I. Andreadis
- Laboratory of Pharmaceutical Technology, School of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Lefkothea C. Papadopoulou
- Laboratory of Pharmacology, School of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Dimitrios G. Fatouros
- Laboratory of Pharmaceutical Technology, School of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
10
|
Stevens B, Winters A, Gutman JA, Fullerton A, Hemenway G, Schatz D, Miltgen N, Wei Q, Abbasi T, Vali S, Singh NK, Drusbosky L, Cogle CR, Hammes A, Abbott D, Jordan CT, Smith C, Pollyea DA. Sequential azacitidine and lenalidomide for patients with relapsed and refractory acute myeloid leukemia: Clinical results and predictive modeling using computational analysis. Leuk Res 2019; 81:43-49. [PMID: 31009835 DOI: 10.1016/j.leukres.2019.04.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 04/04/2019] [Accepted: 04/08/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND Patients with relapsed and refractory (R/R) acute myeloid leukemia (AML) have limited treatment options. Genomically-defined personalized therapies are only applicable for a minority of patients. Therapies without identifiable targets can be effective but patient selection is challenging. The sequential combination of azacitidine with high-dose lenalidomide has shown activity; we aimed to determine the efficacy of this genomically-agnostic regimen in patients with R/R AML, with the intention of applying sophisticated methods to predict responders. METHODS Thirty-seven R/R AML/myelodysplastic syndrome patients were enrolled in a phase 2 study of azacitidine with lenalidomide. The primary endpoint was complete remission (CR) and CR with incomplete blood count recovery (CRi) rate. A computational biological modeling (CBM) approach was applied retrospectively to predict outcomes based on the understood mechanisms of azacitidine and lenalidomide in the setting of each patients' disease. FINDINGS Four of 37 patients (11%) had a CR/CRi; the study failed to meet the alternative hypothesis. Significant toxicity was observed in some cases, with three treatment-related deaths and a 30-day mortality rate of 14%. However, the CBM method predicted responses in 83% of evaluable patients, with a positive and negative predictive value of 80% and 89%, respectively. INTERPRETATION Sequential azacitidine and high-dose lenalidomide is effective in a minority of R/R AML patients; it may be possible to predict responders at the time of diagnosis using a CBM approach. More efforts to predict responses in non-targeted therapies should be made, to spare toxicity in patients unlikely to respond and maximize treatments for those with limited options.
Collapse
Affiliation(s)
- Brett Stevens
- University of Colorado Division of Hematology, Aurora, CO, United States
| | - Amanda Winters
- University of Colorado Children's Hospital, Aurora CO, United States
| | - Jonathan A Gutman
- University of Colorado Division of Hematology, Aurora, CO, United States
| | - Aaron Fullerton
- University of Colorado Division of Hematology, Aurora, CO, United States
| | - Gregory Hemenway
- University of Colorado Division of Hematology, Aurora, CO, United States
| | - Derek Schatz
- University of Colorado Division of Hematology, Aurora, CO, United States
| | - Nicholas Miltgen
- University of Colorado Children's Hospital, Aurora CO, United States
| | - Qi Wei
- University of Colorado Children's Hospital, Aurora CO, United States
| | | | | | | | | | | | - Andrew Hammes
- Center for Innovative Design and Analysis, Department of Biostatistics and Informatics, University of Colorado, Aurora, CO, United States
| | - Diana Abbott
- Center for Innovative Design and Analysis, Department of Biostatistics and Informatics, University of Colorado, Aurora, CO, United States
| | - Craig T Jordan
- University of Colorado Division of Hematology, Aurora, CO, United States
| | - Clayton Smith
- University of Colorado Division of Hematology, Aurora, CO, United States
| | - Daniel A Pollyea
- University of Colorado Division of Hematology, Aurora, CO, United States.
| |
Collapse
|
11
|
Predicting response to BET inhibitors using computational modeling: A BEAT AML project study. Leuk Res 2019; 77:42-50. [PMID: 30642575 PMCID: PMC6442457 DOI: 10.1016/j.leukres.2018.11.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 10/26/2018] [Accepted: 11/18/2018] [Indexed: 12/04/2022]
Abstract
Despite advances in understanding the molecular pathogenesis of acute myeloid leukaemia (AML), overall survival rates remain low. The ability to predict treatment response based on individual cancer genomics using computational modeling will aid in the development of novel therapeutics and personalize care. Here, we used a combination of genomics, computational biology modeling (CBM), ex vivo chemosensitivity assay, and clinical data from 100 randomly selected patients in the Beat AML project to characterize AML sensitivity to a bromodomain (BRD) and extra-terminal (BET) inhibitor. Computational biology modeling was used to generate patient-specific protein network maps of activated and inactivated protein pathways translated from each genomic profile. Digital drug simulations of a BET inhibitor (JQ1) were conducted by quantitatively measuring drug effect using a composite AML disease inhibition score. 93% of predicted disease inhibition scores matched the associated ex vivo IC50 value. Sensitivity and specificity of CBM predictions were 97.67%, and 64.29%, respectively. Genomic predictors of response were identified. Patient samples harbouring chromosomal aberrations del(7q) or −7, +8, or del(5q) and somatic mutations causing ERK pathway dysregulation, responded to JQ1 in both in silico and ex vivo assays. This study shows how a combination of genomics, computational modeling and chemosensitivity testing can identify network signatures associating with treatment response and can inform priority populations for future clinical trials of BET inhibitors.
Collapse
|
12
|
Kobayashi SS, Vali S, Kumar A, Singh N, Abbasi T, Sayeski PP. Identification of myeloproliferative neoplasm drug agents via predictive simulation modeling: assessing responsiveness with micro-environment derived cytokines. Oncotarget 2017; 7:35989-36001. [PMID: 27056884 PMCID: PMC5094977 DOI: 10.18632/oncotarget.8540] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 03/10/2016] [Indexed: 01/06/2023] Open
Abstract
Previous studies have shown that the bone marrow micro-environment supports the myeloproliferative neoplasms (MPN) phenotype including via the production of cytokines that can induce resistance to frontline MPN therapies. However, the mechanisms by which this occurs are poorly understood. Moreover, the ability to rapidly identify drug agents that can act as adjuvants to existing MPN frontline therapies is virtually non-existent. Here, using a novel predictive simulation approach, we sought to determine the effect of various drug agents on MPN cell lines, both with and without the micro-environment derived inflammatory cytokines. We first created individual simulation models for two representative MPN cell lines; HEL and SET-2, based on their genomic mutation and copy number variation (CNV) data. Running computational simulations on these virtual cell line models, we identified a synergistic effect of two drug agents on cell proliferation and viability; namely, the Jak2 kinase inhibitor, G6, and the Bcl-2 inhibitor, ABT737. IL-6 did not show any impact on the cells due to the predicted lack of IL-6 signaling within these cells. Interestingly, TNFα increased the sensitivity of the single drug agents and their use in combination while IFNγ decreased the sensitivity. In summary, this study predictively identified two drug agents that reduce MPN cell viability via independent mechanisms that was prospectively validated. Moreover, their efficacy is either potentiated or inhibited, by some of the micro-environment derived cytokines. Lastly, this study has validated the use of this simulation based technology to prospectively determine such responses.
Collapse
Affiliation(s)
- Susumu S Kobayashi
- Department of Medicine, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA, USA
| | | | - Ansu Kumar
- Cellworks Research India Pvt Ltd., Cellworks Group Inc., Bangalore, India
| | - Neeraj Singh
- Cellworks Research India Pvt Ltd., Cellworks Group Inc., Bangalore, India
| | | | - Peter P Sayeski
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL, USA
| |
Collapse
|
13
|
Drusbosky L, Medina C, Martuscello R, Hawkins KE, Chang M, Lamba JK, Vali S, Kumar A, Singh NK, Abbasi T, Sekeres MA, Mallo M, Sole F, Bejar R, Cogle CR. Computational drug treatment simulations on projections of dysregulated protein networks derived from the myelodysplastic mutanome match clinical response in patients. Leuk Res 2017; 52:1-7. [DOI: 10.1016/j.leukres.2016.11.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 11/02/2016] [Accepted: 11/04/2016] [Indexed: 01/19/2023]
|
14
|
Lanzel EA, Paula Gomez Hernandez M, Bates AM, Treinen CN, Starman EE, Fischer CL, Parashar D, Guthmiller JM, Johnson GK, Abbasi T, Vali S, Brogden KA. Predicting PD-L1 expression on human cancer cells using next-generation sequencing information in computational simulation models. Cancer Immunol Immunother 2016; 65:1511-1522. [PMID: 27688163 DOI: 10.1007/s00262-016-1907-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 09/23/2016] [Indexed: 12/31/2022]
Abstract
PURPOSE Interaction of the programmed death-1 (PD-1) co-receptor on T cells with the programmed death-ligand 1 (PD-L1) on tumor cells can lead to immunosuppression, a key event in the pathogenesis of many tumors. Thus, determining the amount of PD-L1 in tumors by immunohistochemistry (IHC) is important as both a diagnostic aid and a clinical predictor of immunotherapy treatment success. Because IHC reactivity can vary, we developed computational simulation models to accurately predict PD-L1 expression as a complementary assay to affirm IHC reactivity. METHODS Multiple myeloma (MM) and oral squamous cell carcinoma (SCC) cell lines were modeled as examples of our approach. Non-transformed cell models were first simulated to establish non-tumorigenic control baselines. Cell line genomic aberration profiles, from next-generation sequencing (NGS) information for MM.1S, U266B1, SCC4, SCC15, and SCC25 cell lines, were introduced into the workflow to create cancer cell line-specific simulation models. Percentage changes of PD-L1 expression with respect to control baselines were determined and verified against observed PD-L1 expression by ELISA, IHC, and flow cytometry on the same cells grown in culture. RESULT The observed PD-L1 expression matched the predicted PD-L1 expression for MM.1S, U266B1, SCC4, SCC15, and SCC25 cell lines and clearly demonstrated that cell genomics play an integral role by influencing cell signaling and downstream effects on PD-L1 expression. CONCLUSION This concept can easily be extended to cancer patient cells where an accurate method to predict PD-L1 expression would affirm IHC results and improve its potential as a biomarker and a clinical predictor of treatment success.
Collapse
Affiliation(s)
- Emily A Lanzel
- Department of Oral Pathology, Radiology and Medicine, College of Dentistry, University of Iowa, Iowa City, IA, USA
| | - M Paula Gomez Hernandez
- Iowa Institute for Oral Health Research, N423 DSB, College of Dentistry, The University of Iowa, 801 Newton Road, Iowa City, IA, 52242, USA
| | - Amber M Bates
- Iowa Institute for Oral Health Research, N423 DSB, College of Dentistry, The University of Iowa, 801 Newton Road, Iowa City, IA, 52242, USA
| | - Christopher N Treinen
- Iowa Institute for Oral Health Research, N423 DSB, College of Dentistry, The University of Iowa, 801 Newton Road, Iowa City, IA, 52242, USA
| | - Emily E Starman
- Iowa Institute for Oral Health Research, N423 DSB, College of Dentistry, The University of Iowa, 801 Newton Road, Iowa City, IA, 52242, USA
| | - Carol L Fischer
- Iowa Institute for Oral Health Research, N423 DSB, College of Dentistry, The University of Iowa, 801 Newton Road, Iowa City, IA, 52242, USA
| | | | - Janet M Guthmiller
- College of Dentistry, University of Nebraska Medical Center, 40th and Holdrege, Lincoln, NE, USA
| | - Georgia K Johnson
- Department of Periodontics, College of Dentistry, The University of Iowa, Iowa City, IA, USA
| | - Taher Abbasi
- Cellworks Research India Ltd, Whitefield, Bangalore, India
- Cellworks Group Inc, 2033 Gateway Place Suite 500, San Jose, CA, USA
| | - Shireen Vali
- Cellworks Research India Ltd, Whitefield, Bangalore, India
- Cellworks Group Inc, 2033 Gateway Place Suite 500, San Jose, CA, USA
| | - Kim A Brogden
- Iowa Institute for Oral Health Research, N423 DSB, College of Dentistry, The University of Iowa, 801 Newton Road, Iowa City, IA, 52242, USA.
- Department of Periodontics, College of Dentistry, The University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
15
|
Paulus A, Ailawadhi S, Chanan-Khan A. Novel therapeutic targets in Waldenstrom macroglobulinemia. Best Pract Res Clin Haematol 2016; 29:216-228. [PMID: 27825468 DOI: 10.1016/j.beha.2016.08.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Accepted: 08/30/2016] [Indexed: 01/04/2023]
Abstract
Understanding of molecular mechanisms that drive Waldenstrom macroglobulinemia (WM) cell survival are rapidly evolving. This review briefly highlights emerging "WM-relevant" targets; for which therapeutic strategies are currently being investigated in preclinical and clinical studies. With the discovery of MYD88L265P signaling and remarkable activity of ibrutinib in WM, other targets within the B-cell receptor pathway are now being focused on for therapeutic intervention. Additional targets which play a role in WM cell survival include TLR7, 8 and 9, proteasome-associated deubiquitinating enzymes (USP14 and UCHL5), XPO1/CRM1 and AURKA. New drugs for established targets are also discussed. Lastly, we spotlight 3 highly innovative WM-specific therapies: MYD88 peptide inhibitors, MYD88L265P-directed immune activation and CD19-directed chimeric antigen receptor T-cell therapy, which are in various stages of development. Indeed, treatment of WM is poised to undergo a paradigm shift in the coming years towards highly disease-driven and more personalized therapeutic modalities with curative intent.
Collapse
Affiliation(s)
- Aneel Paulus
- Mayo Clinic Jacksonville, Department of Cancer Biology and Division of Hematology and Oncology, United States.
| | - Sikander Ailawadhi
- Mayo Clinic Jacksonville, Division of Hematology and Oncology, United States.
| | - Asher Chanan-Khan
- Mayo Clinic Jacksonville, Division of Hematology and Oncology, United States.
| |
Collapse
|
16
|
Kang J, Chong SJF, Ooi VZQ, Vali S, Kumar A, Kapoor S, Abbasi T, Hirpara JL, Loh T, Goh BC, Pervaiz S. Overexpression of Bcl-2 induces STAT-3 activation via an increase in mitochondrial superoxide. Oncotarget 2016; 6:34191-205. [PMID: 26430964 PMCID: PMC4741445 DOI: 10.18632/oncotarget.5763] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 09/07/2015] [Indexed: 01/28/2023] Open
Abstract
We recently reported a novel interaction between Bcl-2 and Rac1 and linked that to the ability of Bcl-2 to induce a pro-oxidant state in cancer cells. To gain further insight into the functional relevance of this interaction, we utilized computer simulation based on the protein pathway dynamic network created by Cellworks Group Inc. STAT3 was identified among targets that positively correlated with Rac1 and/or Bcl-2 expression levels. Validating this, the activation level of STAT3, as marked by p-Tyr705, particularly in the mitochondria, was significantly higher in Bcl-2-overexpressing cancer cells. Bcl-2-induced STAT3 activation was a function of GTP-loaded Rac1 and NADPH oxidase (Nox)-dependent increase in intracellular superoxide (O2•−). Furthermore, ABT199, a BH-3 specific inhibitor of Bcl-2, as well as silencing of Bcl-2 blocked STAT3 phosphorylation. Interestingly, while inhibiting intracellular O2•− blocked STAT3 phosphorylation, transient overexpression of wild type STAT3 resulted in a significant increase in mitochondrial O2•− production, which was rescued by the functional mutants of STAT3 (Y705F). Notably, a strong correlation between the expression and/or phosphorylation of STAT3 and Bcl-2 was observed in primary tissues derived from patients with different sub-sets of B cell lymphoma. These data demonstrate the presence of a functional crosstalk between Bcl-2, Rac1 and activated STAT3 in promoting a permissive redox milieu for cell survival. Results also highlight the potential utility of a signature involving Bcl-2 overexpression, Rac1 activation and STAT3 phosphorylation for stratifying clinical lymphomas based on disease severity and chemoresistance.
Collapse
Affiliation(s)
- Jia Kang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Stephen Jun Fei Chong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Vignette Zi Qi Ooi
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | | | - Ansu Kumar
- Cellworks Research India Private Limited, Bangalore, India
| | - Shweta Kapoor
- Cellworks Research India Private Limited, Bangalore, India
| | | | - Jayshree L Hirpara
- Experimental Therapeutics Program, Cancer Science Institute, Singapore, Singapore
| | - Thomas Loh
- Department of Otolaryngology, National University Healthcare System, Singapore, Singapore
| | - Boon Cher Goh
- Experimental Therapeutics Program, Cancer Science Institute, Singapore, Singapore
| | - Shazib Pervaiz
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore, Singapore.,National University Cancer Institute, NUHS, Singapore, Singapore.,School of Biomedical Sciences, Curtin University, Perth, Australia
| |
Collapse
|
17
|
Yu P, Lin W. Single-cell Transcriptome Study as Big Data. GENOMICS PROTEOMICS & BIOINFORMATICS 2016; 14:21-30. [PMID: 26876720 PMCID: PMC4792842 DOI: 10.1016/j.gpb.2016.01.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 01/09/2016] [Accepted: 01/10/2016] [Indexed: 12/31/2022]
Abstract
The rapid growth of single-cell RNA-seq studies (scRNA-seq) demands efficient data storage, processing, and analysis. Big-data technology provides a framework that facilitates the comprehensive discovery of biological signals from inter-institutional scRNA-seq datasets. The strategies to solve the stochastic and heterogeneous single-cell transcriptome signal are discussed in this article. After extensively reviewing the available big-data applications of next-generation sequencing (NGS)-based studies, we propose a workflow that accounts for the unique characteristics of scRNA-seq data and primary objectives of single-cell studies.
Collapse
Affiliation(s)
- Pingjian Yu
- Genomics and Bioinformatics Lab, Baylor Institute for Immunology Research, Dallas, TX 75204, USA
| | - Wei Lin
- Genomics and Bioinformatics Lab, Baylor Institute for Immunology Research, Dallas, TX 75204, USA.
| |
Collapse
|
18
|
Vizirianakis IS, Mystridis GA, Avgoustakis K, Fatouros DG, Spanakis M. Enabling personalized cancer medicine decisions: The challenging pharmacological approach of PBPK models for nanomedicine and pharmacogenomics (Review). Oncol Rep 2016; 35:1891-904. [PMID: 26781205 DOI: 10.3892/or.2016.4575] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 10/27/2015] [Indexed: 11/05/2022] Open
Abstract
The existing tumor heterogeneity and the complexity of cancer cell biology critically demand powerful translational tools with which to support interdisciplinary efforts aiming to advance personalized cancer medicine decisions in drug development and clinical practice. The development of physiologically based pharmacokinetic (PBPK) models to predict the effects of drugs in the body facilitates the clinical translation of genomic knowledge and the implementation of in vivo pharmacology experience with pharmacogenomics. Such a direction unequivocally empowers our capacity to also make personalized drug dosage scheme decisions for drugs, including molecularly targeted agents and innovative nanoformulations, i.e. in establishing pharmacotyping in prescription. In this way, the applicability of PBPK models to guide individualized cancer therapeutic decisions of broad clinical utility in nanomedicine in real-time and in a cost-affordable manner will be discussed. The latter will be presented by emphasizing the need for combined efforts within the scientific borderlines of genomics with nanotechnology to ensure major benefits and productivity for nanomedicine and personalized medicine interventions.
Collapse
Affiliation(s)
- Ioannis S Vizirianakis
- Laboratory of Pharmacology, Department of Pharmaceutical Sciences, School of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki GR‑54124, Greece
| | - George A Mystridis
- Laboratory of Pharmacology, Department of Pharmaceutical Sciences, School of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki GR‑54124, Greece
| | - Konstantinos Avgoustakis
- Laboratory of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Patras, Patras GR-26504, Greece
| | - Dimitrios G Fatouros
- Laboratory of Pharmaceutical Technology, Department of Pharmaceutical Sciences, Aristotle University of Thessaloniki, Thessaloniki GR-54124, Greece
| | - Marios Spanakis
- Computational BioMedicine Laboratory, Institute of Computer Science, Foundation for Research and Technology-Hellas, Heraklion GR-71110, Crete, Greece
| |
Collapse
|
19
|
Sarin H. Conserved molecular mechanisms underlying the effects of small molecule xenobiotic chemotherapeutics on cells. Mol Clin Oncol 2015; 4:326-368. [PMID: 26998284 DOI: 10.3892/mco.2015.714] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 12/08/2015] [Indexed: 12/14/2022] Open
Abstract
For proper determination of the apoptotic potential of chemoxenobiotics in synergism, it is important to understand the modes, levels and character of interactions of chemoxenobiotics with cells in the context of predicted conserved biophysical properties. Chemoxenobiotic structures are studied with respect to atom distribution over molecular space, the predicted overall octanol-to-water partition coefficient (Log OWPC; unitless) and molecular size viz a viz van der Waals diameter (vdWD). The Log OWPC-to-vdWD (nm-1 ) parameter is determined, and where applicable, hydrophilic interacting moiety/core-to-vdWD (nm-1 ) and lipophilic incorporating hydrophobic moiety/core-to-vdWD (nm-1 ) parameters of their part-structures are determined. The cellular and sub-cellular level interactions of the spectrum of xenobiotic chemotherapies have been characterized, for which a classification system has been developed based on predicted conserved biophysical properties with respect to the mode of chemotherapeutic effect. The findings of this study are applicable towards improving the effectiveness of existing combination chemotherapy regimens and the predictive accuracy of personalized cancer treatment algorithms as well as towards the selection of appropriate novel xenobiotics with the potential to be potent chemotherapeutics for dendrimer nanoparticle-based effective transvascular delivery.
Collapse
Affiliation(s)
- Hemant Sarin
- Freelance Investigator in Translational Science and Medicine, Charleston, WV 25314, USA
| |
Collapse
|