1
|
Butner JD, Martin GV, Wang Z, Corradetti B, Ferrari M, Esnaola N, Chung C, Hong DS, Welsh JW, Hasegawa N, Mittendorf EA, Curley SA, Chen SH, Pan PY, Libutti SK, Ganesan S, Sidman RL, Pasqualini R, Arap W, Koay EJ, Cristini V. Early prediction of clinical response to checkpoint inhibitor therapy in human solid tumors through mathematical modeling. eLife 2021; 10:70130. [PMID: 34749885 PMCID: PMC8629426 DOI: 10.7554/elife.70130] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 10/25/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Checkpoint inhibitor therapy of cancer has led to markedly improved survival of a subset of patients in multiple solid malignant tumor types, yet the factors driving these clinical responses or lack thereof are not known. We have developed a mechanistic mathematical model for better understanding these factors and their relations in order to predict treatment outcome and optimize personal treatment strategies. Methods: Here, we present a translational mathematical model dependent on three key parameters for describing efficacy of checkpoint inhibitors in human cancer: tumor growth rate (α), tumor-immune infiltration (Λ), and immunotherapy-mediated amplification of anti-tumor response (µ). The model was calibrated by fitting it to a compiled clinical tumor response dataset (n = 189 patients) obtained from published anti-PD-1 and anti-PD-L1 clinical trials, and then validated on an additional validation cohort (n = 64 patients) obtained from our in-house clinical trials. Results: The derived parameters Λ and µ were both significantly different between responding versus nonresponding patients. Of note, our model appropriately classified response in 81.4% of patients by using only tumor volume measurements and within 2 months of treatment initiation in a retrospective analysis. The model reliably predicted clinical response to the PD-1/PD-L1 class of checkpoint inhibitors across multiple solid malignant tumor types. Comparison of model parameters to immunohistochemical measurement of PD-L1 and CD8+ T cells confirmed robust relationships between model parameters and their underlying biology. Conclusions: These results have demonstrated reliable methods to inform model parameters directly from biopsy samples, which are conveniently obtainable as early as the start of treatment. Together, these suggest that the model parameters may serve as early and robust biomarkers of the efficacy of checkpoint inhibitor therapy on an individualized per-patient basis. Funding: We gratefully acknowledge support from the Andrew Sabin Family Fellowship, Center for Radiation Oncology Research, Sheikh Ahmed Center for Pancreatic Cancer Research, GE Healthcare, Philips Healthcare, and institutional funds from the University of Texas M.D. Anderson Cancer Center. We have also received Cancer Center Support Grants from the National Cancer Institute (P30CA016672 to the University of Texas M.D. Anderson Cancer Center and P30CA072720 the Rutgers Cancer Institute of New Jersey). This research has also been supported in part by grants from the National Science Foundation Grant DMS-1930583 (ZW, VC), the National Institutes of Health (NIH) 1R01CA253865 (ZW, VC), 1U01CA196403 (ZW, VC), 1U01CA213759 (ZW, VC), 1R01CA226537 (ZW, RP, WA, VC), 1R01CA222007 (ZW, VC), U54CA210181 (ZW, VC), and the University of Texas System STARS Award (VC). BC acknowledges support through the SER Cymru II Programme, funded by the European Commission through the Horizon 2020 Marie Skłodowska-Curie Actions (MSCA) COFUND scheme and the Welsh European Funding Office (WEFO) under the European Regional Development Fund (ERDF). EK has also received support from the Project Purple, NIH (U54CA210181, U01CA200468, and U01CA196403), and the Pancreatic Cancer Action Network (16-65-SING). MF was supported through NIH/NCI center grant U54CA210181, R01CA222959, DoD Breast Cancer Research Breakthrough Level IV Award W81XWH-17-1-0389, and the Ernest Cockrell Jr. Presidential Distinguished Chair at Houston Methodist Research Institute. RP and WA received serial research awards from AngelWorks, the Gillson-Longenbaugh Foundation, and the Marcus Foundation. This work was also supported in part by grants from the National Cancer Institute to SHC (R01CA109322, R01CA127483, R01CA208703, and U54CA210181 CITO pilot grant) and to PYP (R01CA140243, R01CA188610, and U54CA210181 CITO pilot grant). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
Collapse
Affiliation(s)
- Joseph D Butner
- The Houston Methodist Research Institute, Houston, United States
| | - Geoffrey V Martin
- The University of Texas MD Anderson Cancer Center, Houston, United States
| | - Zhihui Wang
- The Houston Methodist Research Institute, Houston, United States
| | - Bruna Corradetti
- The Houston Methodist Research Institute, Houston, United States
| | - Mauro Ferrari
- The Houston Methodist Research Institute, Houston, United States
| | - Nestor Esnaola
- The Houston Methodist Research Institute, Houston, United States
| | - Caroline Chung
- The University of Texas MD Anderson Cancer Center, Houston, United States
| | - David S Hong
- The University of Texas MD Anderson Cancer Center, Houston, United States
| | - James W Welsh
- The Houston Methodist Research Institute, Houston, United States
| | - Naomi Hasegawa
- University of Texas Health Science Center, Houston, United States
| | | | | | - Shu-Hsia Chen
- The Houston Methodist Research Institute, Houston, United States
| | - Ping-Ying Pan
- The Houston Methodist Research Institute, Houston, United States
| | | | | | - Richard L Sidman
- Department of Neurology, Harvard Medical School, Boston, United States
| | | | - Wadih Arap
- Hematology and Oncology, Rutgers Cancer Institute of New Jersey, Newark, United States
| | - Eugene J Koay
- University of Texas MD Anderson Cancer Center, Houston, United States
| | | |
Collapse
|
2
|
Moradi Kashkooli F, Soltani M. Evaluation of solid tumor response to sequential treatment cycles via a new computational hybrid approach. Sci Rep 2021; 11:21475. [PMID: 34728726 PMCID: PMC8563754 DOI: 10.1038/s41598-021-00989-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 10/21/2021] [Indexed: 12/22/2022] Open
Abstract
The development of an in silico approach that evaluates and identifies appropriate treatment protocols for individuals could help grow personalized treatment and increase cancer patient lifespans. With this motivation, the present study introduces a novel approach for sequential treatment cycles based on simultaneously examining drug delivery, tumor growth, and chemotherapy efficacy. This model incorporates the physical conditions of tumor geometry, including tumor, capillary network, and normal tissue assuming real circumstances, as well as the intravascular and interstitial fluid flow, drug concentration, chemotherapy efficacy, and tumor recurrence. Three treatment approaches-maximum tolerated dose (MTD), metronomic chemotherapy (MC), and chemo-switching (CS)-as well as different chemotherapy schedules are investigated on a real tumor geometry extracted from image. Additionally, a sensitivity analysis of effective parameters of drug is carried out to evaluate the potential of using different other drugs in cancer treatment. The main findings are: (i) CS, MC, and MTD have the best performance in reducing tumor cells, respectively; (ii) multiple doses raise the efficacy of drugs that have slower clearance, higher diffusivity, and lower to medium binding affinities; (iii) the suggested approach to eradicating tumors is to reduce their cells to a predetermined rate through chemotherapy and then apply adjunct therapy.
Collapse
Affiliation(s)
| | - M Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran.
- Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, ON, Canada.
- Cancer Biology Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran, Iran.
- Advanced Bioengineering Initiative Center, Computational Medicine Center, K. N. Toosi University of Technology, Tehran, Iran.
| |
Collapse
|
3
|
Amero P, Lokesh GLR, Chaudhari RR, Cardenas-Zuniga R, Schubert T, Attia YM, Montalvo-Gonzalez E, Elsayed AM, Ivan C, Wang Z, Cristini V, Franciscis VD, Zhang S, Volk DE, Mitra R, Rodriguez-Aguayo C, Sood AK, Lopez-Berestein G. Conversion of RNA Aptamer into Modified DNA Aptamers Provides for Prolonged Stability and Enhanced Antitumor Activity. J Am Chem Soc 2021; 143:7655-7670. [PMID: 33988982 DOI: 10.1021/jacs.9b10460] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Aptamers, synthetic single-strand oligonucleotides that are similar in function to antibodies, are promising as therapeutics because of their minimal side effects. However, the stability and bioavailability of the aptamers pose a challenge. We developed aptamers converted from RNA aptamer to modified DNA aptamers that target phospho-AXL with improved stability and bioavailability. On the basis of the comparative analysis of a library of 17 converted modified DNA aptamers, we selected aptamer candidates, GLB-G25 and GLB-A04, that exhibited the highest bioavailability, stability, and robust antitumor effect in in vitro experiments. Backbone modifications such as thiophosphate or dithiophosphate and a covalent modification of the 5'-end of the aptamer with polyethylene glycol optimized the pharmacokinetic properties, improved the stability of the aptamers in vivo by reducing nuclease hydrolysis and renal clearance, and achieved high and sustained inhibition of AXL at a very low dose. Treatment with these modified aptamers in ovarian cancer orthotopic mouse models significantly reduced tumor growth and the number of metastases. This effective silencing of the phospho-AXL target thus demonstrated that aptamer specificity and bioavailability can be improved by the chemical modification of existing aptamers for phospho-AXL. These results lay the foundation for the translation of these aptamer candidates and companion biomarkers to the clinic.
Collapse
Affiliation(s)
- Paola Amero
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Ganesh L R Lokesh
- McGovern Medical School, Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center, Houston, Texas 77030, United States
| | - Rajan R Chaudhari
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Roberto Cardenas-Zuniga
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | | | - Yasmin M Attia
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States.,Pharmacology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Kasr Al Eini Street, Fom El Khalig, Cairo 11796, Egypt
| | - Efigenia Montalvo-Gonzalez
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States.,Integral Laboratory of Food Research, Technological Institute of Tepic, Avenue Tecnologico 2595, 63175 Tepic, Nayarit Mexico
| | - Abdelrahman M Elsayed
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Cairo 11675, Egypt
| | - Cristina Ivan
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Zhihui Wang
- Mathematics in Medicine Program, The Houston Methodist Research Institute, 6670 Bertner Ave, Houston, Texas 77030, United States
| | - Vittorio Cristini
- Mathematics in Medicine Program, The Houston Methodist Research Institute, 6670 Bertner Ave, Houston, Texas 77030, United States
| | - Vittorio de Franciscis
- Istituto di Endocrinologia ed Oncologia Sperimentale, CNR, 80131 Naples, Italy.,National Research Council (CNR), Institute of Genetic and Biomedical Research (IRGB)-UOS Milan via Rita Levi Montalcini, 20090 Pieve Emanuele (MI), Italy.,Harvard Medical School Initiative for RNA Medicine, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Shuxing Zhang
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - David E Volk
- McGovern Medical School, Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center, Houston, Texas 77030, United States
| | - Rahul Mitra
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Cristian Rodriguez-Aguayo
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States.,Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Anil K Sood
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States.,Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States.,Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Gabriel Lopez-Berestein
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States.,Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States.,Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| |
Collapse
|
4
|
Anaya DA, Dogra P, Wang Z, Haider M, Ehab J, Jeong DK, Ghayouri M, Lauwers GY, Thomas K, Kim R, Butner JD, Nizzero S, Ramírez JR, Plodinec M, Sidman RL, Cavenee WK, Pasqualini R, Arap W, Fleming JB, Cristini V. A Mathematical Model to Estimate Chemotherapy Concentration at the Tumor-Site and Predict Therapy Response in Colorectal Cancer Patients with Liver Metastases. Cancers (Basel) 2021; 13:cancers13030444. [PMID: 33503971 PMCID: PMC7866038 DOI: 10.3390/cancers13030444] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 01/21/2021] [Indexed: 12/22/2022] Open
Abstract
Simple Summary It is known that drug transport barriers in the tumor determine drug concentration at the tumor site, causing disparity from the systemic (plasma) drug concentration. However, current clinical standard of care still bases dosage and treatment optimization on the systemic concentration of drugs. Here, we present a proof of concept observational cohort study to accurately estimate drug concentration at the tumor site from mathematical modeling using biologic, clinical, and imaging/perfusion data, and correlate it with outcome in colorectal cancer liver metastases. We demonstrate that drug concentration at the tumor site, not in systemic circulation, can be used as a credible biomarker for predicting chemotherapy outcome, and thus our mathematical modeling approach can be applied prospectively in the clinic to personalize treatment design to optimize outcome. Abstract Chemotherapy remains a primary treatment for metastatic cancer, with tumor response being the benchmark outcome marker. However, therapeutic response in cancer is unpredictable due to heterogeneity in drug delivery from systemic circulation to solid tumors. In this proof-of-concept study, we evaluated chemotherapy concentration at the tumor-site and its association with therapy response by applying a mathematical model. By using pre-treatment imaging, clinical and biologic variables, and chemotherapy regimen to inform the model, we estimated tumor-site chemotherapy concentration in patients with colorectal cancer liver metastases, who received treatment prior to surgical hepatic resection with curative-intent. The differential response to therapy in resected specimens, measured with the gold-standard Tumor Regression Grade (TRG; from 1, complete response to 5, no response) was examined, relative to the model predicted systemic and tumor-site chemotherapy concentrations. We found that the average calculated plasma concentration of the cytotoxic drug was essentially equivalent across patients exhibiting different TRGs, while the estimated tumor-site chemotherapeutic concentration (eTSCC) showed a quadratic decline from TRG = 1 to TRG = 5 (p < 0.001). The eTSCC was significantly lower than the observed plasma concentration and dropped by a factor of ~5 between patients with complete response (TRG = 1) and those with no response (TRG = 5), while the plasma concentration remained stable across TRG groups. TRG variations were driven and predicted by differences in tumor perfusion and eTSCC. If confirmed in carefully planned prospective studies, these findings will form the basis of a paradigm shift in the care of patients with potentially curable colorectal cancer and liver metastases.
Collapse
Affiliation(s)
- Daniel A. Anaya
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (M.H.); (J.E.); (R.K.); (J.B.F.)
- Correspondence: (D.A.A.); (V.C.); Tel.: +1-813-745-1432 (D.A.A.); +1-505-934-1813 (V.C.); Fax: +1-813-745-7229 (D.A.A.)
| | - Prashant Dogra
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX 77030, USA; (P.D.); (Z.W.); (J.D.B.); (S.N.); (J.R.R.)
| | - Zhihui Wang
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX 77030, USA; (P.D.); (Z.W.); (J.D.B.); (S.N.); (J.R.R.)
| | - Mintallah Haider
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (M.H.); (J.E.); (R.K.); (J.B.F.)
| | - Jasmina Ehab
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (M.H.); (J.E.); (R.K.); (J.B.F.)
| | - Daniel K. Jeong
- Department of Diagnostic Imaging and Interventional Radiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (D.K.J.); (M.G.); (G.Y.L.); (K.T.)
| | - Masoumeh Ghayouri
- Department of Diagnostic Imaging and Interventional Radiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (D.K.J.); (M.G.); (G.Y.L.); (K.T.)
| | - Gregory Y. Lauwers
- Department of Diagnostic Imaging and Interventional Radiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (D.K.J.); (M.G.); (G.Y.L.); (K.T.)
| | - Kerry Thomas
- Department of Diagnostic Imaging and Interventional Radiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (D.K.J.); (M.G.); (G.Y.L.); (K.T.)
| | - Richard Kim
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (M.H.); (J.E.); (R.K.); (J.B.F.)
| | - Joseph D. Butner
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX 77030, USA; (P.D.); (Z.W.); (J.D.B.); (S.N.); (J.R.R.)
| | - Sara Nizzero
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX 77030, USA; (P.D.); (Z.W.); (J.D.B.); (S.N.); (J.R.R.)
| | - Javier Ruiz Ramírez
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX 77030, USA; (P.D.); (Z.W.); (J.D.B.); (S.N.); (J.R.R.)
| | - Marija Plodinec
- Biozentrum and the Swiss Nanoscience Institute & ARTIDIS AG, University of Basel, 4056 Basel, Switzerland;
| | - Richard L. Sidman
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA;
| | - Webster K. Cavenee
- Ludwig Institute for Cancer Research, University of California-San Diego, La Jolla, CA 92093, USA;
| | - Renata Pasqualini
- Rutgers Cancer Institute of New Jersey & Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103, USA;
| | - Wadih Arap
- Rutgers Cancer Institute of New Jersey & Division of Hematology/Oncology, Department of Medicine Rutgers New Jersey Medical School, Newark, NJ 07103, USA;
| | - Jason B. Fleming
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (M.H.); (J.E.); (R.K.); (J.B.F.)
| | - Vittorio Cristini
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX 77030, USA; (P.D.); (Z.W.); (J.D.B.); (S.N.); (J.R.R.)
- Correspondence: (D.A.A.); (V.C.); Tel.: +1-813-745-1432 (D.A.A.); +1-505-934-1813 (V.C.); Fax: +1-813-745-7229 (D.A.A.)
| |
Collapse
|
5
|
Gorur A, Bayraktar R, Ivan C, Mokhlis HA, Bayraktar E, Kahraman N, Karakas D, Karamil S, Kabil NN, Kanlikilicer P, Aslan B, Tamer L, Wang Z, Cristini V, Lopez-Berestein G, Calin G, Ozpolat B. ncRNA therapy with miRNA-22-3p suppresses the growth of triple-negative breast cancer. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 23:930-943. [PMID: 33614241 PMCID: PMC7868999 DOI: 10.1016/j.omtn.2021.01.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 01/14/2021] [Indexed: 12/14/2022]
Abstract
Deregulation of noncoding RNAs, including microRNAs (miRs), is implicated in the pathogenesis of many human cancers, including breast cancer. Through extensive analysis of The Cancer Genome Atlas, we found that expression of miR-22-3p is markedly lower in triple-negative breast cancer (TNBC) than in normal breast tissue. The restoration of miR-22-3p expression led to significant inhibition of TNBC cell proliferation, colony formation, migration, and invasion. We demonstrated that miR-22-3p reduces eukaryotic elongation factor 2 kinase (eEF2K) expression by directly binding to the 3' untranslated region of eEF2K mRNA. Inhibition of EF2K expression recapitulated the effects of miR-22-3p on TNBC cell proliferation, motility, invasion, and suppression of phosphatidylinositol 3-kinase/Akt and Src signaling. Systemic administration of miR-22-3p in single-lipid nanoparticles significantly suppressed tumor growth in orthotopic MDA-MB-231 and MDA-MB-436 TNBC models. Evaluation of the tumor response, following miR-22-3p therapy in these models using a novel mathematical model factoring in various in vivo parameters, demonstrated that the therapy is highly effective against TNBC. These findings suggest that miR-22-3p functions as a tumor suppressor by targeting clinically significant oncogenic pathways and that miR-22-3p loss contributes to TNBC growth and progression. The restoration of miR-22-3p expression is a potential novel noncoding RNA-based therapy for TNBC.
Collapse
Affiliation(s)
- Aysegul Gorur
- Department of Experimental Therapeutics, Unit 1950, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA.,Department of Biochemistry, School of Medicine, Mersin University, Mersin, Turkey
| | - Recep Bayraktar
- Department of Experimental Therapeutics, Unit 1950, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Cristina Ivan
- Department of Experimental Therapeutics, Unit 1950, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA.,Center for RNA Interference and Non-Coding RNAs, Unit 2080, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Hamada Ahmed Mokhlis
- Department of Experimental Therapeutics, Unit 1950, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, The University of Al-Azhar, Cairo, Egypt
| | - Emine Bayraktar
- Department of Experimental Therapeutics, Unit 1950, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Nermin Kahraman
- Department of Experimental Therapeutics, Unit 1950, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Didem Karakas
- Department of Experimental Therapeutics, Unit 1950, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Selda Karamil
- Department of Experimental Therapeutics, Unit 1950, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Nashwa N Kabil
- Department of Experimental Therapeutics, Unit 1950, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Pinar Kanlikilicer
- Department of Experimental Therapeutics, Unit 1950, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Burcu Aslan
- Department of Experimental Therapeutics, Unit 1950, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Lulufer Tamer
- Department of Biochemistry, School of Medicine, Mersin University, Mersin, Turkey
| | - Zhihui Wang
- Mathematics in Medicine, Houston Methodist Research Institute, 6565 Fannin Street, Houston, TX 77030, USA
| | - Vittorio Cristini
- Mathematics in Medicine, Houston Methodist Research Institute, 6565 Fannin Street, Houston, TX 77030, USA
| | - Gabriel Lopez-Berestein
- Department of Experimental Therapeutics, Unit 1950, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA.,Center for RNA Interference and Non-Coding RNAs, Unit 2080, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - George Calin
- Department of Experimental Therapeutics, Unit 1950, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA.,Center for RNA Interference and Non-Coding RNAs, Unit 2080, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Bulent Ozpolat
- Department of Experimental Therapeutics, Unit 1950, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA.,Center for RNA Interference and Non-Coding RNAs, Unit 2080, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| |
Collapse
|
6
|
Machiraju GB, Mallick P, Frieboes HB. Multicompartment modeling of protein shedding kinetics during vascularized tumor growth. Sci Rep 2020; 10:16709. [PMID: 33028917 PMCID: PMC7542472 DOI: 10.1038/s41598-020-73866-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 09/10/2020] [Indexed: 02/07/2023] Open
Abstract
Identification of protein biomarkers for cancer diagnosis and prognosis remains a critical unmet clinical need. A major reason is that the dynamic relationship between proliferating and necrotic cell populations during vascularized tumor growth, and the associated extra- and intra-cellular protein outflux from these populations into blood circulation remains poorly understood. Complementary to experimental efforts, mathematical approaches have been employed to effectively simulate the kinetics of detectable surface proteins (e.g., CA-125) shed into the bloodstream. However, existing models can be difficult to tune and may be unable to capture the dynamics of non-extracellular proteins, such as those shed from necrotic and apoptosing cells. The models may also fail to account for intra-tumoral spatial and microenvironmental heterogeneity. We present a new multi-compartment model to simulate heterogeneously vascularized growing tumors and the corresponding protein outflux. Model parameters can be tuned from histology data, including relative vascular volume, mean vessel diameter, and distance from vasculature to necrotic tissue. The model enables evaluating the difference in shedding rates between extra- and non-extracellular proteins from viable and necrosing cells as a function of heterogeneous vascularization. Simulation results indicate that under certain conditions it is possible for non-extracellular proteins to have superior outflux relative to extracellular proteins. This work contributes towards the goal of cancer biomarker identification by enabling simulation of protein shedding kinetics based on tumor tissue-specific characteristics. Ultimately, we anticipate that models like the one introduced herein will enable examining origins and circulating dynamics of candidate biomarkers, thus facilitating marker selection for validation studies.
Collapse
Affiliation(s)
- Gautam B Machiraju
- Biomedical Informatics Training Program, Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Parag Mallick
- Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, CA, USA.
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA.
| | - Hermann B Frieboes
- Department of Bioengineering, University of Louisville, Louisville, KY, USA.
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA.
- Center for Predictive Medicine, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
7
|
Dogra P, Chuang YL, Butner JD, Cristini V, Wang Z. Development of a Physiologically-Based Mathematical Model for Quantifying Nanoparticle Distribution in Tumors. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2020; 2019:2852-2855. [PMID: 31946487 DOI: 10.1109/embc.2019.8856503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Nanomedicine holds promise for the treatment of cancer, as it enables tumor-targeted drug delivery. However, reports on translation of most nanomedicine strategies to the clinic so far have been less than satisfactory, in part due to insufficient understanding of the effects of nanoparticle (NP) physiochemical properties and physiological variables on their pharmacological behavior. In this paper, we present a multiscale mathematical model to examine the efficacy of NP delivery to solid tumors; as a case example, we apply the model to a clinically detectable primary pancreatic ductal adenocarcinoma (PDAC) to assess tissue-scale spatiotemporal distribution profiles of NPs. We integrate NP systemic disposition kinetics with NP-cell interactions in PDAC abstractly described as a two-dimensional structure, which is then parameterized with human physiological data obtained from published literature. Through model analysis of delivery efficiency, we verify the multiscale approach by showing that NP concentration kinetics of interest in various compartments predicted by the whole-body scale model were in agreement with those obtained from the tissue-scale model. We also found that more NPs were trapped in the outer well-perfused tumor region than the inner semi-necrotic domain. Further development of the model may provide a useful tool for optimal NP design and physiological interventions.
Collapse
|
8
|
Dogra P, Ramírez JR, Peláez MJ, Wang Z, Cristini V, Parasher G, Rawat M. Mathematical Modeling to Address Challenges in Pancreatic Cancer. Curr Top Med Chem 2020; 20:367-376. [PMID: 31893993 PMCID: PMC7279939 DOI: 10.2174/1568026620666200101095641] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 09/10/2019] [Accepted: 10/20/2019] [Indexed: 12/30/2022]
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) is regarded as one of the most lethal cancer types for its challenges associated with early diagnosis and resistance to standard chemotherapeutic agents, thereby leading to a poor five-year survival rate. The complexity of the disease calls for a multidisciplinary approach to better manage the disease and improve the status quo in PDAC diagnosis, prognosis, and treatment. To this end, the application of quantitative tools can help improve the understanding of disease mechanisms, develop biomarkers for early diagnosis, and design patient-specific treatment strategies to improve therapeutic outcomes. However, such approaches have only been minimally applied towards the investigation of PDAC, and we review the current status of mathematical modeling works in this field.
Collapse
Affiliation(s)
- Prashant Dogra
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Javier Ruiz Ramírez
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - María J. Peláez
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX 77030, USA
- Applied Physics Graduate Program, Rice University, Houston, TX 77005, USA
| | - Zhihui Wang
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Vittorio Cristini
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Gulshan Parasher
- Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Manmeet Rawat
- Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| |
Collapse
|
9
|
Abstract
Cancer continues to be among the leading healthcare problems worldwide, and efforts continue not just to find better drugs, but also better drug delivery methods. The need for delivering cytotoxic agents selectively to cancerous cells, for improved safety and efficacy, has triggered the application of nanotechnology in medicine. This effort has provided drug delivery systems that can potentially revolutionize cancer treatment. Nanocarriers, due to their capacity for targeted drug delivery, can shift the balance of cytotoxicity from healthy to cancerous cells. The field of cancer nanomedicine has made significant progress, but challenges remain that impede its clinical translation. Several biophysical barriers to the transport of nanocarriers to the tumor exist, and a much deeper understanding of nano-bio interactions is necessary to change the status quo. Mathematical modeling has been instrumental in improving our understanding of the physicochemical and physiological underpinnings of nanomaterial behavior in biological systems. Here, we present a comprehensive review of literature on mathematical modeling works that have been and are being employed towards a better understanding of nano-bio interactions for improved tumor delivery efficacy.
Collapse
|
10
|
Brocato TA, Brown-Glaberman U, Wang Z, Selwyn RG, Wilson CM, Wyckoff EF, Lomo LC, Saline JL, Hooda-Nehra A, Pasqualini R, Arap W, Brinker CJ, Cristini V. Predicting breast cancer response to neoadjuvant chemotherapy based on tumor vascular features in needle biopsies. JCI Insight 2019; 5:126518. [PMID: 30835256 DOI: 10.1172/jci.insight.126518] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
In clinical breast cancer intervention, selection of the optimal treatment protocol based on predictive biomarkers remains an elusive goal. Here, we present a modeling tool to predict the likelihood of breast cancer response to neoadjuvant chemotherapy using patient specific tumor vasculature biomarkers. A semi-automated analysis was implemented and performed on 3990 histological images from 48 patients, with 10-208 images analyzed for each patient. We applied a histology-based model to resected primary breast cancer tumors (n = 30), and then evaluated a cohort of patients (n = 18) undergoing neoadjuvant chemotherapy, collecting pre- and post-treatment pathology specimens and MRI data. We found that core biopsy samples can be used with acceptable accuracy (r = 0.76) to determine histological parameters representative of the whole tissue region. Analysis of model histology parameters obtained from tumor vasculature measurements, specifically diffusion distance divided by radius of drug source (L/rb) and blood volume fraction (BVF), provides a statistically significant separation of patients obtaining a pathologic complete response (pCR) from those that do not (Student's t-test; P < 0.05). With this model, it is feasible to evaluate primary breast tumor vasculature biomarkers in a patient specific manner, thereby allowing a precision approach to breast cancer treatment.
Collapse
Affiliation(s)
- Terisse A Brocato
- Department of Chemical and Biological Engineering and Center for Biomedical Engineering, University of New Mexico, Albuquerque, New Mexico, USA
| | - Ursa Brown-Glaberman
- University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico, USA
| | - Zhihui Wang
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA.,Department of Imaging Physics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Reed G Selwyn
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,Department of Radiology, University of New Mexico, Albuquerque, New Mexico, USA
| | - Colin M Wilson
- Department of Radiology, University of New Mexico, Albuquerque, New Mexico, USA
| | - Edward F Wyckoff
- Center for Micro-Engineered Materials, University of New Mexico, Albuquerque, New Mexico, USA
| | - Lesley C Lomo
- Department of Pathology, University of Utah, Salt Lake City, Utah, USA
| | - Jennifer L Saline
- Department of Radiology, University of New Mexico, Albuquerque, New Mexico, USA
| | - Anupama Hooda-Nehra
- Rutgers Cancer Institute of New Jersey, Newark, New Jersey, USA.,Division of Hematology/Oncology, Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Renata Pasqualini
- Rutgers Cancer Institute of New Jersey, Newark, New Jersey, USA.,Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Wadih Arap
- Rutgers Cancer Institute of New Jersey, Newark, New Jersey, USA.,Division of Hematology/Oncology, Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - C Jeffrey Brinker
- Department of Chemical and Biological Engineering and Center for Biomedical Engineering, University of New Mexico, Albuquerque, New Mexico, USA.,Center for Micro-Engineered Materials, University of New Mexico, Albuquerque, New Mexico, USA.,Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA.,Self-Assembled Materials Department, Sandia National Laboratories, Albuquerque, New Mexico, USA
| | - Vittorio Cristini
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA.,Department of Imaging Physics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Department of Nanomedicine, Methodist Hospital Research Institute, Houston, Texas, USA
| |
Collapse
|
11
|
Wang Z, Deisboeck TS. Dynamic Targeting in Cancer Treatment. Front Physiol 2019; 10:96. [PMID: 30890944 PMCID: PMC6413712 DOI: 10.3389/fphys.2019.00096] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 01/25/2019] [Indexed: 12/18/2022] Open
Abstract
With the advent of personalized medicine, design and development of anti-cancer drugs that are specifically targeted to individual or sets of genes or proteins has been an active research area in both academia and industry. The underlying motivation for this approach is to interfere with several pathological crosstalk pathways in order to inhibit or at the very least control the proliferation of cancer cells. However, after initially conferring beneficial effects, if sub-lethal, these artificial perturbations in cell function pathways can inadvertently activate drug-induced up- and down-regulation of feedback loops, resulting in dynamic changes over time in the molecular network structure and potentially causing drug resistance as seen in clinics. Hence, the targets or their combined signatures should also change in accordance with the evolution of the network (reflected by changes to the structure and/or functional output of the network) over the course of treatment. This suggests the need for a "dynamic targeting" strategy aimed at optimizing tumor control by interfering with different molecular targets, at varying stages. Understanding the dynamic changes of this complex network under various perturbed conditions due to drug treatment is extremely challenging under experimental conditions let alone in clinical settings. However, mathematical modeling can facilitate studying these effects at the network level and beyond, and also accelerate comparison of the impact of different dosage regimens and therapeutic modalities prior to sizeable investment in risky and expensive clinical trials. A dynamic targeting strategy based on the use of mathematical modeling can be a new, exciting research avenue in the discovery and development of therapeutic drugs.
Collapse
Affiliation(s)
- Zhihui Wang
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, United States.,Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Thomas S Deisboeck
- Department of Radiology, Harvard-MIT (HST) Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| |
Collapse
|
12
|
Chamseddine IM, Frieboes HB, Kokkolaras M. Design Optimization of Tumor Vasculature-Bound Nanoparticles. Sci Rep 2018; 8:17768. [PMID: 30538267 PMCID: PMC6290012 DOI: 10.1038/s41598-018-35675-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 10/26/2018] [Indexed: 01/07/2023] Open
Abstract
Nanotherapy may constitute a promising approach to target tumors with anticancer drugs while minimizing systemic toxicity. Computational modeling can enable rapid evaluation of nanoparticle (NP) designs and numerical optimization. Here, an optimization study was performed using an existing tumor model to find NP size and ligand density that maximize tumoral NP accumulation while minimizing tumor size. Optimal NP avidity lies at lower bound of feasible values, suggesting reduced ligand density to prolong NP circulation. For the given set of tumor parameters, optimal NP diameters were 288 nm to maximize NP accumulation and 334 nm to minimize tumor diameter, leading to uniform NP distribution and adequate drug load. Results further show higher dependence of NP biodistribution on the NP design than on tumor morphological parameters. A parametric study with respect to drug potency was performed. The lower the potency of the drug, the bigger the difference is between the maximizer of NP accumulation and the minimizer of tumor size, indicating the existence of a specific drug potency that minimizes the differential between the two optimal solutions. This study shows the feasibility of applying optimization to NP designs to achieve efficacious cancer nanotherapy, and offers a first step towards a quantitative tool to support clinical decision making.
Collapse
Affiliation(s)
| | - Hermann B Frieboes
- Department of Bioengineering, University of Louisville, Louisville, KY, USA
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Michael Kokkolaras
- Department of Mechanical Engineering, McGill University, Montreal, Quebec, Canada.
- GERAD - Group for Research in Decision Analysis, Montreal, Quebec, Canada.
| |
Collapse
|
13
|
Dogra P, Adolphi NL, Wang Z, Lin YS, Butler KS, Durfee PN, Croissant JG, Noureddine A, Coker EN, Bearer EL, Cristini V, Brinker CJ. Establishing the effects of mesoporous silica nanoparticle properties on in vivo disposition using imaging-based pharmacokinetics. Nat Commun 2018; 9:4551. [PMID: 30382084 PMCID: PMC6208419 DOI: 10.1038/s41467-018-06730-z] [Citation(s) in RCA: 174] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 09/13/2018] [Indexed: 12/31/2022] Open
Abstract
The progress of nanoparticle (NP)-based drug delivery has been hindered by an inability to establish structure-activity relationships in vivo. Here, using stable, monosized, radiolabeled, mesoporous silica nanoparticles (MSNs), we apply an integrated SPECT/CT imaging and mathematical modeling approach to understand the combined effects of MSN size, surface chemistry and routes of administration on biodistribution and clearance kinetics in healthy rats. We show that increased particle size from ~32- to ~142-nm results in a monotonic decrease in systemic bioavailability, irrespective of route of administration, with corresponding accumulation in liver and spleen. Cationic MSNs with surface exposed amines (PEI) have reduced circulation, compared to MSNs of identical size and charge but with shielded amines (QA), due to rapid sequestration into liver and spleen. However, QA show greater total excretion than PEI and their size-matched neutral counterparts (TMS). Overall, we provide important predictive functional correlations to support the rational design of nanomedicines.
Collapse
Affiliation(s)
- Prashant Dogra
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Natalie L Adolphi
- Department of Biochemistry and Molecular Biology, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Zhihui Wang
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Department of Imaging Physics, University of Texas MD Anderson Cancer Center, Houston, TX, 78701, USA
| | - Yu-Shen Lin
- Department of Internal Medicine, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Kimberly S Butler
- Center for Micro-Engineered Materials, University of New Mexico, Albuquerque, NM, 87131, USA
- Chemical and Biological Engineering, University of New Mexico, Albuquerque, NM, 87131, USA
- Sandia National Laboratories, Department of Nanobiology, Albuquerque, NM, 87123, USA
| | - Paul N Durfee
- Center for Micro-Engineered Materials, University of New Mexico, Albuquerque, NM, 87131, USA
- Cancer Research and Treatment Center, Molecular Genetics and Microbiology, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Jonas G Croissant
- Center for Micro-Engineered Materials, University of New Mexico, Albuquerque, NM, 87131, USA
- Chemical and Biological Engineering, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Achraf Noureddine
- Center for Micro-Engineered Materials, University of New Mexico, Albuquerque, NM, 87131, USA
- Chemical and Biological Engineering, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Eric N Coker
- Sandia National Laboratories, Applied Optical and Plasma Science, Albuquerque, NM, 87185, USA
| | - Elaine L Bearer
- Department of Pathology, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Vittorio Cristini
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, 77030, USA.
- Department of Imaging Physics, University of Texas MD Anderson Cancer Center, Houston, TX, 78701, USA.
| | - C Jeffrey Brinker
- Center for Micro-Engineered Materials, University of New Mexico, Albuquerque, NM, 87131, USA.
- Chemical and Biological Engineering, University of New Mexico, Albuquerque, NM, 87131, USA.
- Cancer Research and Treatment Center, Molecular Genetics and Microbiology, University of New Mexico, Albuquerque, NM, 87131, USA.
- Sandia National Laboratories, Self-Assembled Materials Department, Albuquerque, NM, 87185, USA.
| |
Collapse
|
14
|
Hamis S, Nithiarasu P, Powathil GG. What does not kill a tumour may make it stronger: In silico insights into chemotherapeutic drug resistance. J Theor Biol 2018; 454:253-267. [DOI: 10.1016/j.jtbi.2018.06.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 06/04/2018] [Accepted: 06/12/2018] [Indexed: 12/01/2022]
|
15
|
Understanding the Connection between Nanoparticle Uptake and Cancer Treatment Efficacy using Mathematical Modeling. Sci Rep 2018; 8:7538. [PMID: 29795392 PMCID: PMC5967303 DOI: 10.1038/s41598-018-25878-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 04/23/2018] [Indexed: 12/12/2022] Open
Abstract
Nanoparticles have shown great promise in improving cancer treatment efficacy while reducing toxicity and treatment side effects. Predicting the treatment outcome for nanoparticle systems by measuring nanoparticle biodistribution has been challenging due to the commonly unmatched, heterogeneous distribution of nanoparticles relative to free drug distribution. We here present a proof-of-concept study that uses mathematical modeling together with experimentation to address this challenge. Individual mice with 4T1 breast cancer were treated with either nanoparticle-delivered or free doxorubicin, with results demonstrating improved cancer kill efficacy of doxorubicin loaded nanoparticles in comparison to free doxorubicin. We then developed a mathematical theory to render model predictions from measured nanoparticle biodistribution, as determined using graphite furnace atomic absorption. Model analysis finds that treatment efficacy increased exponentially with increased nanoparticle accumulation within the tumor, emphasizing the significance of developing new ways to optimize the delivery efficiency of nanoparticles to the tumor microenvironment.
Collapse
|
16
|
Curtis LT, van Berkel VH, Frieboes HB. Pharmacokinetic/pharmacodynamic modeling of combination-chemotherapy for lung cancer. J Theor Biol 2018; 448:38-52. [PMID: 29614265 DOI: 10.1016/j.jtbi.2018.03.035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Revised: 03/23/2018] [Accepted: 03/26/2018] [Indexed: 02/06/2023]
Abstract
Chemotherapy for non-small cell lung cancer (NSCLC) typically involves a doublet regimen for a number of cycles. For any particular patient, a course of treatment is usually chosen from a large number of combinational protocols with drugs in concomitant or sequential administration. In spite of newer drugs and protocols, half of patients with early disease will live less than five years and 95% of those with advanced disease survive for less than one year. Here, we apply mathematical modeling to simulate tumor response to multiple drug regimens, with the capability to assess maximum tolerated dose (MTD) as well as metronomic drug administration. We couple pharmacokinetic-pharmacodynamic intracellular multi-compartment models with a model of vascularized tumor growth, setting input parameters from in vitro data, and using the models to project potential response in vivo. This represents an initial step towards the development of a comprehensive virtual system to evaluate tumor response to combinatorial drug regimens, with the goal to more efficiently identify optimal course of treatment with patient tumor-specific data. We evaluate cisplatin and gemcitabine with clinically-relevant dosages, and simulate four treatment NSCLC scenarios combining MTD and metronomic therapy. This work thus establishes a framework for systematic evaluation of tumor response to combination chemotherapy. The results with the chosen parameter set indicate that although a metronomic regimen may provide advantage over MTD, the combination of these regimens may not necessarily offer improved response. Future model evaluation of chemotherapy possibilities may help to assess their potential value to obtain sustained NSCLC regression for particular patients, with the ultimate goal of optimizing multiple-drug chemotherapy regimens in clinical practice.
Collapse
Affiliation(s)
- Louis T Curtis
- Department of Bioengineering, University of Louisville, Lutz Hall 419, Louisville, KY 40208, USA
| | - Victor H van Berkel
- Department of Cardiovascular and Thoracic Surgery, University of Louisville, KY, USA; James Graham Brown Cancer Center, University of Louisville, KY, USA
| | - Hermann B Frieboes
- Department of Bioengineering, University of Louisville, Lutz Hall 419, Louisville, KY 40208, USA; James Graham Brown Cancer Center, University of Louisville, KY, USA; Department of Pharmacology & Toxicology, University of Louisville, KY, USA.
| |
Collapse
|
17
|
Nargis NN, Aldredge RC, Guy RD. The influence of soluble fragments of extracellular matrix (ECM) on tumor growth and morphology. Math Biosci 2017; 296:1-16. [PMID: 29208360 DOI: 10.1016/j.mbs.2017.11.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 11/22/2017] [Accepted: 11/30/2017] [Indexed: 01/27/2023]
Abstract
A major challenge in matrix-metalloproteinase (MMP) target validation and MMP-inhibitor-drug development for anti-cancer clinical trials is to better understand their complex roles (often competing with each other) in tumor progression. While there is extensive research on the growth-promoting effects of MMPs, the growth-inhibiting effects of MMPs has not been investigated thoroughly. So we develop a continuum model of tumor growth and invasion including chemotaxis and haptotaxis in order to examine the complex interaction between the tumor and its host microenvironment and to explore the inhibiting influence of the gradients of soluble fragments of extracellular matrix (ECM) density on tumor growth and morphology. Previously, it was shown both computationally (in one spatial dimension) and experimentally that the chemotactic pull due to soluble ECM gradients is anti-invasive, contrary to the traditional view of the role of chemotaxis in malignant invasion [1]. With two-dimensional numerical simulation and using a level set based tumor-host interface capturing method, we examine the effects of chemotaxis on the progression and morphology of a tumor growing in nutrient-rich and nutrient-poor microenvironments which was not investigated before. In particular we examine how the geometry of the growing tumor is affected when placed in different environments. We also investigate the effects of varying ECM degradation rate, the production rate of matrix degrading enzymes (MDE), and the conversion of ECM into soluble ECM. We find that chemotaxis due to ECM-fragment gradients strongly influences tumor growth and morphology, and that the instabilities caused by tumor cell proliferation and haptotactic movements can be prevented if chemotaxis is sufficiently strong. The influence of chemotaxis and the above factors on tumor growth and morphology are found to be more prominent in nutrient-poor environments than in nutrient-rich environments. So we extend our investigations of these antinvasive chemotactic influences by examining the effects of cell-cell and cell-ECM adhesion and low proliferation rate for tumors growing in low-nutrient environments. We find that as the extent of chemotaxis increases, the effects of adhesion on tumor growth and shape become negligible. Under conditions of low cell mitosis, chemotaxis may cause the tumor to shrink, as the extent of chemotaxis increases. Both stable and unstable tumor shrinkage are predicted by our model. Unexpectedly, in some cases chemotaxis may contribute toward developing instability where haptotaxis alone induces stable growth.
Collapse
Affiliation(s)
- Nurun N Nargis
- Department of Mechanical and Aerospace Engineering, University of California, Davis, CA 95616, USA
| | - Ralph C Aldredge
- Department of Mechanical and Aerospace Engineering, University of California, Davis, CA 95616, USA.
| | - Robert D Guy
- Department of Mathematics, University of California, Davis, CA 95616, USA
| |
Collapse
|
18
|
Tennill TA, Gross ME, Frieboes HB. Automated analysis of co-localized protein expression in histologic sections of prostate cancer. PLoS One 2017; 12:e0178362. [PMID: 28552967 PMCID: PMC5446169 DOI: 10.1371/journal.pone.0178362] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 05/11/2017] [Indexed: 12/13/2022] Open
Abstract
An automated approach based on routinely-processed, whole-slide immunohistochemistry (IHC) was implemented to study co-localized protein expression in tissue samples. Expression of two markers was chosen to represent stromal (CD31) and epithelial (Ki-67) compartments in prostate cancer. IHC was performed on whole-slide sections representing low-, intermediate-, and high-grade disease from 15 patients. The automated workflow was developed using a training set of regions-of-interest in sequential tissue sections. Protein expression was studied on digital representations of IHC images across entire slides representing formalin-fixed paraffin embedded blocks. Using the training-set, the known association between Ki-67 and Gleason grade was confirmed. CD31 expression was more heterogeneous across samples and remained invariant with grade in this cohort. Interestingly, the Ki-67/CD31 ratio was significantly increased in high (Gleason ≥ 8) versus low/intermediate (Gleason ≤7) samples when assessed in the training-set and the whole-tissue block images. Further, the feasibility of the automated approach to process Tissue Microarray (TMA) samples in high throughput was evaluated. This work establishes an initial framework for automated analysis of co-localized protein expression and distribution in high-resolution digital microscopy images based on standard IHC techniques. Applied to a larger sample population, the approach may help to elucidate the biologic basis for the Gleason grade, which is the strongest, single factor distinguishing clinically aggressive from indolent prostate cancer.
Collapse
Affiliation(s)
- Thomas A. Tennill
- Department of Bioengineering, University of Louisville, Louisville, KY, United States of America
| | - Mitchell E. Gross
- Lawrence J. Elliston Institute for Transformational Medicine, University of Southern California, Los Angeles, CA, United States of America
| | - Hermann B. Frieboes
- Department of Bioengineering, University of Louisville, Louisville, KY, United States of America
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States of America
| |
Collapse
|
19
|
Abstract
In vivo imaging, which enables us to peer deeply within living subjects, is producing tremendous opportunities both for clinical diagnostics and as a research tool. Contrast material is often required to clearly visualize the functional architecture of physiological structures. Recent advances in nanomaterials are becoming pivotal to generate the high-resolution, high-contrast images needed for accurate, precision diagnostics. Nanomaterials are playing major roles in imaging by delivering large imaging payloads, yielding improved sensitivity, multiplexing capacity, and modularity of design. Indeed, for several imaging modalities, nanomaterials are now not simply ancillary contrast entities, but are instead the original and sole source of image signal that make possible the modality's existence. We address the physicochemical makeup/design of nanomaterials through the lens of the physical properties that produce contrast signal for the cognate imaging modality-we stratify nanomaterials on the basis of their (i) magnetic, (ii) optical, (iii) acoustic, and/or (iv) nuclear properties. We evaluate them for their ability to provide relevant information under preclinical and clinical circumstances, their in vivo safety profiles (which are being incorporated into their chemical design), their modularity in being fused to create multimodal nanomaterials (spanning multiple different physical imaging modalities and therapeutic/theranostic capabilities), their key properties, and critically their likelihood to be clinically translated.
Collapse
Affiliation(s)
- Bryan Ronain Smith
- Stanford University , 3155 Porter Drive, #1214, Palo Alto, California 94304-5483, United States
| | - Sanjiv Sam Gambhir
- The James H. Clark Center , 318 Campus Drive, First Floor, E-150A, Stanford, California 94305-5427, United States
| |
Collapse
|
20
|
Frieboes HB, Curtis LT, Wu M, Kani K, Mallick P. Simulation of the Protein-Shedding Kinetics of a Fully Vascularized Tumor. Cancer Inform 2015; 14:163-75. [PMID: 26715830 PMCID: PMC4687979 DOI: 10.4137/cin.s35374] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 11/09/2015] [Accepted: 11/15/2015] [Indexed: 12/12/2022] Open
Abstract
Circulating biomarkers are of significant interest for cancer detection and treatment personalization. However, the biophysical processes that determine how proteins are shed from cancer cells or their microenvironment, diffuse through tissue, enter blood vasculature, and persist in circulation remain poorly understood. Since approaches primarily focused on experimental evaluation are incapable of measuring the shedding and persistence for every possible marker candidate, we propose an interdisciplinary computational/experimental approach that includes computational modeling of tumor tissue heterogeneity. The model implements protein production, transport, and shedding based on tumor vascularization, cell proliferation, hypoxia, and necrosis, thus quantitatively relating the tumor and circulating proteomes. The results highlight the dynamics of shedding as a function of protein diffusivity and production. Linking the simulated tumor parameters to clinical tumor and vascularization measurements could potentially enable this approach to reveal the tumor-specific conditions based on the protein detected in circulation and thus help to more accurately manage cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Hermann B Frieboes
- Department of Bioengineering, University of Louisville, Louisville, KY, USA. ; James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Louis T Curtis
- Department of Bioengineering, University of Louisville, Louisville, KY, USA
| | - Min Wu
- Department of Engineering Sciences and Applied Mathematics, Northwestern University, Chicago, IL, USA
| | - Kian Kani
- Center for Applied Molecular Medicine, University of Southern California, Los Angeles, CA, USA
| | - Parag Mallick
- Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, CA, USA
| |
Collapse
|