Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Mehran Makvandi is active.

Publication


Featured researches published by Mehran Makvandi.


Cancer Research | 2016

A Radiotracer Strategy to Quantify PARP-1 Expression In Vivo Provides a Biomarker That Can Enable Patient Selection for PARP Inhibitor Therapy.

Mehran Makvandi; Kuiying Xu; Brian P. Lieberman; Redmond-Craig Anderson; Samuel Sander Effron; Harrison D. Winters; Chenbo Zeng; Elizabeth S. McDonald; Daniel A. Pryma; Roger A. Greenberg; Robert H. Mach

Despite the availability of PARP inhibitors for cancer therapy, a biomarker to clearly stratify patients for selection of this treatment remains lacking. Here we describe a radiotracer-based method that addresses this issue, using the novel compound [(125)I] KX1: as a PARP-1-selective radiotracer that can accurately measure PARP-1 expression in vitro and in vivo The pharmacologic properties of the PARP radiotracer [(125)I] KX1: was characterized in multiple cell lines where single-agent sensitivity was correlated with [(125)I] KX1: binding to PARP-1. In vivo evaluation of [(125)I] KX1: verified in vitro results, validating PARP radiotracers to define PARP-1 enzyme expression as an in vivo biomarker. Notably, PARP-1 expression as quantified by [(125)I] KX1: correlated positively with the cytotoxic sensitivity of cell lines evaluated with PARP inhibitors. Overall, our results defined a novel technology with the potential to serve as a companion diagnostic to identify patients most likely to respond therapeutically to a PARP inhibitor. Cancer Res; 76(15); 4516-24. ©2016 AACR.


JCI insight | 2017

A patient-derived-xenograft platform to study BRCA-deficient ovarian cancers

Erin George; Hyoung Kim; Clemens Krepler; Brandon Wenz; Mehran Makvandi; Janos L. Tanyi; Eric J. Brown; Rugang Zhang; Patricia Brafford; Stephanie Jean; Robert H. Mach; Yiling Lu; Gordon B. Mills; Meenhard Herlyn; Mark A. Morgan; Xiaochen Zhang; Robert A. Soslow; Ronny Drapkin; Neil Johnson; Ying Zheng; George Cotsarelis; Katherine L. Nathanson; Fiona Simpkins

Approximately 50% of high-grade serous ovarian cancers (HGSOCs) have defects in genes involved in homologous recombination (HR) (i.e., BRCA1/2). Preclinical models to optimize therapeutic strategies for HR-deficient (HRD) HGSOC are lacking. We developed a preclinical platform for HRD HGSOCs that includes primary tumor cultures, patient-derived xenografts (PDXs), and molecular imaging. Models were characterized by immunohistochemistry, targeted sequencing, and reverse-phase protein array analysis. We also tested PDX tumor response to PARP, CHK1, and ATR inhibitors. Fourteen orthotopic HGSOC PDX models with BRCA mutations (BRCAMUT) were established with a 93% success rate. The orthotopic PDX model emulates the natural progression of HGSOC, including development of a primary ovarian tumor and metastasis to abdominal viscera. PDX response to standard chemotherapy correlated to that demonstrated in the patient. Pathogenic mutations and HGSOC markers were preserved after multiple mouse passages, indicating retention of underlying molecular mechanisms of carcinogenesis. A BRCA2MUT PDX with high p-CHK1 demonstrated a similar delay of tumor growth in response to PARP, CHK1, and ATR inhibitors. A poly (ADP-ribose) polymerase (PARP) inhibitor radiotracer correlated with PARP1 activity and showed response to PARP inhibition in the BRCA2MUT PDX model. In summary, the orthotopic HGSOC PDX represents a robust and reliable model to optimize therapeutic strategies for BRCAMUT HGSOC.


Nuclear Medicine and Biology | 2016

The pre-clinical characterization of an alpha-emitting sigma-2 receptor targeted radiotherapeutic.

Mehran Makvandi; Brian P. Lieberman; Ben LeGeyt; Catherine Hou; David A. Mankoff; Robert H. Mach; Daniel A. Pryma

RATIONALE The sigma-2 receptor is a protein with a Heme binding region and is capable of receptor-mediated endocytosis. It is overexpressed in many cancers making it a potential vector for therapeutic drug delivery. Our objective was to introduce an alpha-emitting radionuclide, astatine-211, into a selective sigma-2 ligand moiety to provide cytotoxic capabilities without adversely altering the pharmacological characteristics. In this study we investigated the in vitro/in vivo tumor targeting and estimated dosimetry of alpha-emitting sigma-2 ligand, 5-(astato-(211)At)-N-(4-(6,7-dimethoxy-3,4-dihydroisoquinolin-2(1H)-yl)butyl)-2,3-dimethoxybenzamide ((211)At-MM3), in a pre-clinical human breast cancer model. METHODS Astatine-211 was produced in a cyclotron and isolated by dry distillation. Radiosynthesis of (211)At-MM3 was performed using a tin precursor through radioastatodestannylation. In vitro sigma-2 binding experiments using (211)At-MM3 were carried out in live EMT6 and MDA-MB-231 breast cancer cells and liver homogenate tissue. In vivo biodistribution experiments were performed using EMT6 mouse breast cancer cells in BALB/c female mice. Approximately 370 kBq of (211)At-MM3 was administered intravenously and at time points of 5 min, 1, 2, 4, 8, and 24 h organs/tissue were harvested. Estimated human dosimetry was extrapolated from biodistribution data using OLINDA/EXM (VU e-Innovations). RESULTS Astatine-211 was successfully produced and isolated in quantities suitable for in vitro and small animal in vivo experiments. Radiosynthesis of (211)At-MM3 was reproducible with high radiochemical purity. Astatine-211-MM3 exhibited picomolar affinity to the sigma-2 receptor in contrast to the iodinated analog that had nanomolar affinity. Prolonged tumor targeting was measured through biodistribution studies with a maximal tumor to muscle ratio of 9.02 at 4h. Estimated human dosimetry revealed doses of up to 370 MBq in an adult female patient were below organ radiation limits with the potential to provide a high therapeutic dose to tumors. CONCLUSION The sigma-2 receptor could serve as a suitable targeting platform for designing radiotherapeutics. (211)At-MM3 showed tumor targeting properties in vitro/in vivo and favorable estimated human dosimetry establishing the proof of concept for future development as a radiotherapeutic for the treatment of breast cancer.


Nuclear Medicine and Biology | 2016

Iodinated benzimidazole PARP radiotracer for evaluating PARP1/2 expression in vitro and in vivo

Redmond-Craig Anderson; Mehran Makvandi; Kuiying Xu; Brian P. Lieberman; Chenbo Zeng; Daniel A. Pryma; Robert H. Mach

BACKGROUND PARP inhibitors (PARPi) have the potential to impact cancer therapy in a selective patient population; however, despite current patient selection methods clinical trials have shown mixed response rates. It is therefore clinically useful to determine which patients will respond prior to receiving PARPi therapy. One essential biomarker is to measure the level of PARP enzyme expression in tumors. Small molecule radiotracers have been developed to accurately quantify PARP-1 expression in vitro and in vivo. [125I]KX-02-019 is the first report of a radioiodinated analogue of the benzimidazole class of PARPi. Herein, we studied the pharmacological properties of [125I]KX-02-019 as well as the in vivo biodistribution. METHODS [125I]KX-02-019 was evaluated in both cancer and non-cancer cell lines. We evaluated the pharmacologic properties of [125I]KX-02-019 in live cells by measuring enzyme association and dissociation kinetics, saturation, and specificity. In addition, competitive inhibition experiments were carried out with commercially available PARPi. Protein expression was analyzed by Western blot to compare PARP-1 and PARP-2 expression across cell lines studied. The biodistribution was studied in a mouse EMT6 tumor model at time points of 0.5, 1, 2, 4 and 6h. RESULTS [125I]KX-02-019 showed subtle differences in pharmacological properties in the absence of PARP-2. In addition, [125I]KX-02-019 was competitively displaced by clinical PARPi. In vivo biodistribution studies showed an increasing tumor to muscle ratio over 6h as well as fast clearance from healthy tissues. CONCLUSION [125I]KX-02-019 has binding sites in both PARP1 KO cells as well as PARP2 KO cells showing higher affinity for PARP-2. This observation is supported by a decrease in binding affinity in PARP2 KO cells compared to PARP1 KO cells. The pharmacologic and biological properties of [125I]KX-02-019 studied in vitro and in vivo showed that this analogue may be useful in determining pharmacokinetic and pharmacodynamic properties of clinical PARPi.


Journal of Clinical Investigation | 2018

A PET imaging agent for evaluating PARP-1 expression in ovarian cancer

Mehran Makvandi; Austin Pantel; Lauren Schwartz; Erin K. Schubert; Kuiying Xu; Chia-Ju Hsieh; Catherine Hou; Hyoung Kim; Chi-Chang Weng; Harrison D. Winters; Robert K. Doot; Michael D. Farwell; Daniel A. Pryma; Roger A. Greenberg; David A. Mankoff; Fiona Simpkins; Robert H. Mach; Lilie L. Lin

BACKGROUND. Poly(ADP-ribose) polymerase (PARP) inhibitors are effective in a broad population of patients with ovarian cancer; however, resistance caused by low enzyme expression of the drug target PARP-1 remains to be clinically evaluated in this context. We hypothesize that PARP-1 expression is variable in ovarian cancer and can be quantified in primary and metastatic disease using a novel PET imaging agent. METHODS. We used a translational approach to describe the significance of PET imaging of PARP-1 in ovarian cancer. First, we produced PARP1-KO ovarian cancer cell lines using CRISPR/Cas9 gene editing to test the loss of PARP-1 as a resistance mechanism to all clinically used PARP inhibitors. Next, we performed preclinical microPET imaging studies using ovarian cancer patient–derived xenografts in mouse models. Finally, in a phase I PET imaging clinical trial we explored PET imaging as a regional marker of PARP-1 expression in primary and metastatic disease through correlative tissue histology. RESULTS. We found that deletion of PARP1 causes resistance to all PARP inhibitors in vitro, and microPET imaging provides proof of concept as an approach to quantify PARP-1 in vivo. Clinically, we observed a spectrum of standard uptake values (SUVs) ranging from 2–12 for PARP-1 in tumors. In addition, we found a positive correlation between PET SUVs and fluorescent immunohistochemistry for PARP-1 (r2 = 0.60). CONCLUSION. This work confirms the translational potential of a PARP-1 PET imaging agent and supports future clinical trials to test PARP-1 expression as a method to stratify patients for PARP inhibitor therapy. TRIAL REGISTRATION. Clinicaltrials.gov NCT02637934. FUNDING. Research reported in this publication was supported by the Department of Defense OC160269, a Basser Center team science grant, NIH National Cancer Institute R01CA174904, a Department of Energy training grant DE-SC0012476, Abramson Cancer Center Radiation Oncology pilot grants, the Marsha Rivkin Foundation, Kaleidoscope of Hope Foundation, and Paul Calabresi K12 Career Development Award 5K12CA076931.


Targeted Oncology | 2018

Alpha-Emitters and Targeted Alpha Therapy in Oncology: from Basic Science to Clinical Investigations

Mehran Makvandi; Edouard Dupis; Jonathan W. Engle; F. Meiring Nortier; Michael E. Fassbender; Sam Simon; Eva R. Birnbaum; Robert Atcher; Kevin D. John; Olivier Rixe; Jeffrey P. Norenberg

Alpha-emitters are radionuclides that decay through the emission of high linear energy transfer α-particles and possess favorable pharmacologic profiles for cancer treatment. When coupled with monoclonal antibodies, peptides, small molecules, or nanoparticles, the excellent cytotoxic capability of α-particle emissions has generated a strong interest in exploring targeted α-therapy in the pre-clinical setting and more recently in clinical trials in oncology. Multiple obstacles have been overcome by researchers and clinicians to accelerate the development of targeted α-therapies, especially with the recent improvement in isotope production and purification, but also with the development of innovative strategies for optimized targeting. Numerous studies have demonstrated the in vitro and in vivo efficacy of the targeted α-therapy. Radium-223 (223Ra) dichloride (Xofigo®) is the first α-emitter to have received FDA approval for the treatment of prostate cancer with metastatic bone lesions. There is a significant increase in the number of clinical trials in oncology using several radionuclides such as Actinium-225 (225Ac), Bismuth-213 (213Bi), Lead-212 (212Pb), Astatine (211At) or Radium-223 (223Ra) assessing their safety and preliminary activity. This review will cover their therapeutic application as well as summarize the investigations that provide the foundation for further clinical development.


Organic Letters | 2018

Rapid Cu-Catalyzed [211At]Astatination and [125I]Iodination of Boronic Esters at Room Temperature

Sean W. Reilly; Mehran Makvandi; Kuiying Xu; Robert H. Mach

Access to 211At- and 125I-radiolabeled compounds in excellent RCCs and RCYs was achieved in just 10 min at room temperature using a Cu catalyst. The reaction conditions are applicable to a broad class of aryl and heteroaryl boronic reagents with varying steric and electronic properties as well as late-stage astatination and iodination of anticancer PARP inhibitors. This protocol eliminates the traditional need for toxic organotin reagents, elevated temperatures, and extended reaction times, providing a more practical and environmentally friendly approach to developing α-emitting radiotherapeutics.


Drug Discovery Today: Technologies | 2017

Inflammation and DNA damage: Probing pathways to cancer and neurodegeneration

Mehran Makvandi; Mark A. Sellmyer; Robert H. Mach

Cancer and neurodegeneration represent two opposite ends of the biological spectrum but contain many common biological mechanisms. Two such mechanisms include the elevated levels of oxidative stress and DNA damage. In this brief review, we describe current approaches for imaging these biological pathways with the molecular imaging technique, Positron Emission Tomography (PET), and the potential of PET imaging studies to measure the efficacy of anticancer drugs and strategies for delaying the progression of neurodegenerative disorders.


Cancer Research | 2017

Abstract 5197: Targeting PARP-1 to deliver alpha-particles to cancer chromatin

Laura N. Puentes; Kuiying Xu; Catherine Hou; Robert H. Mach; John M. Maris; Daniel A. Pryma; Mehran Makvandi

Introduction: Neuroblastoma (NB) is a radiosensitive pediatric cancer that develops in the sympathetic nervous system and typically affects children under the age of 10. High-risk NB is associated with a 40% 5-year survival rate. Nuclear enzyme poly (ADP-ribose) polymerase 1 (PARP-1) is overexpressed in high-risk NB, making it an attractive target for alpha-particle therapy. Alpha-particles have a short path length and high linear energy transfer, causing dense ionizations across DNA, inducing double stranded breaks that result in cell death. The purpose of this study was to explore a newly developed radiotherapeutic ([ 211 At]MM4) that combines the targeting potential of a small molecule PARP inhibitor (PARPi) with the cytotoxic effects of 211 At in high-risk NB. Methods: In vitro cytotoxicity was performed in a panel of high-risk NB cell lines to evaluate the relative potency of [ 211 At]MM4. Next, DNA damage was assessed by measuring gH2AX foci formation at 1, 4, and 24 hrs after [ 211 At]MM4 treatment. In parallel, PARP-1 expression was measured in response to therapy and cleaved PARP-1 was quantified to assess apoptosis. Cell cycle analysis was performed after treatment to identify therapy related effects. The in-vivo biodistribution of [ 211 At]MM4 was performed alongside ex-vivo autoradiography. Tumor cytology for PARP-1, gH2AX, and Ki-67 was performed in response to therapy. Finally, in-vivo therapy experiments were performed. Results: Cytotoxicity data indicated a significant reduction in cell viability following treatment for all six NB cell lines, in-vitro co-incubation studies confirmed the specificity of [ 211 At]-MM4 to its drug target. Immunofluorescence analysis showed a dose-dependent increase in both gH2AX and PARP-1 expression. The in-vivo biodistribution of [ 211 At]MM4 revealed rapid tumor targeting at 1 hr and clearance from all tissues at 4 hrs. Ex-vivo autoradiography showed a tumor-muscle ratio greater than 6. Tumor cytology revealed DNA damage measured by gH2AX and PARP-1 expression increased following treatment. Small colonies of proliferating tumor cells were detected after treatment using Ki-67 staining. In-vivo therapy efficacy studies revealed low fractionated doses were tolerable and resulted in significant delay in tumor regrowth. Conclusion: [ 211 At]MM4 is a novel alpha-emitting radiotherapeutic that specifically targets nuclear PARP-1 overexpression in neuroblastoma and incites double-stranded breaks in cancer DNA. The cytotoxic effects of [ 211 At]MM4 have been experimentally validated both in-vitro and in-vivo; the results of these experiments confirm the therapeutic potential of [ 211 At]MM4 as a viable treatment option for high-risk neuroblastoma. Citation Format: Laura Puentes, Kuiying Xu, Catherine Hou, Robert H. Mach, John M. Maris, Daniel A. Pryma, Mehran Makvandi. Targeting PARP-1 to deliver alpha-particles to cancer chromatin [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 5197. doi:10.1158/1538-7445.AM2017-5197


Nuclear Medicine and Biology | 2016

Comparative evaluation of 4 and 6-carbon spacer conformationally flexible tetrahydroisoquinolinyl benzamide analogues for imaging the sigma-2 receptor status of solid tumors

Iljung Lee; Brian P. Lieberman; Shihong Li; Catherine Hou; Mehran Makvandi; Robert H. Mach

INTRODUCTION Nine novel analogues were synthesized including a 6-carbon spacer analogue of ISO-1 (7). They have moderate binding affinity for sigma-2 (σ2) receptors and high selectivity for σ2 receptors relative to sigma-1 (σ1) receptors. METHODS ([18F]7) was synthesized and evaluated as a candidate ligand for positron emission (PET) imaging of the σ2 receptor in tumors. Radioligand [18F]7 was radiolabeled with 18F via displacement of the corresponding mesylate precursor with [18F]fluoride. Cellular uptake study of [18F]7 was performed in EMT-6 tumor cell, and in vivo biodistribution study of [18F]7 and microPET imaging study of [18F]3 and [18F]7 carried out in female Balb/c mice bearing EMT-6 tumors. RESULTS [18F]7 had a respectable tumor uptake (1.55%ID/g at 60min post-injection) and high tumor/muscle ratios at 60 and 120min post-injection. MicroPET imaging of [18F]7 in tumor-bearing mice as above showed significant tumor localization and a high tumor/muscle ratio as well. CONCLUSIONS These results are similar to or better than [18F]ISO-1 ([18F]3), which indicates that [18F]7 has potential for imaging the σ2 receptor status of solid tumors.

Collaboration


Dive into the Mehran Makvandi's collaboration.

Top Co-Authors

Avatar

Robert H. Mach

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Daniel A. Pryma

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Catherine Hou

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Kuiying Xu

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chenbo Zeng

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

David A. Mankoff

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

John M. Maris

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Fiona Simpkins

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge