Network


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

Hotspot


Dive into the research topics where Kelly Mojica is active.

Publication


Featured researches published by Kelly Mojica.


The Journal of Nuclear Medicine | 2012

A Vaccinia Virus Encoding the Human Sodium Iodide Symporter Facilitates Long-Term Image Monitoring of Virotherapy and Targeted Radiotherapy of Pancreatic Cancer

Dana Haddad; Pat Zanzonico; Sean Carlin; Chun-Hao Chen; Nanhai G. Chen; Qian Zhang; Yong A. Yu; Valerie A. Longo; Kelly Mojica; Richard J. Aguilar; Aladar A. Szalay; Yuman Fong

To assess therapeutic response and potential toxicity of oncolytic virotherapy, a noninvasive, deep-tissue imaging modality is needed. This study aimed to assess the feasibility, parameters, and determining factors of serial imaging and long-term monitoring of virotherapy and the therapeutic response of pancreatic cancer xenografts treated with a vaccinia virus carrying the human sodium iodide symporter GLV-1h153. Methods: Pancreatic cancer xenografts (PANC-1) in nude mice were treated systemically or intratumorally with GLV-1h153 and serially imaged using 124I PET at 1, 2, 3, and 5 wk after viral injection. Signal intensity was compared with tumor therapeutic response and optical imaging, and tumors were histologically analyzed for morphology and the presence of virus. Autoradiography was performed using technetium-pertechnetate and γ-scintigraphy to assess determining factors for radiouptake in tumors. Finally, the enhanced therapeutic effect of combination therapy with GLV-1h153 and systemic radioiodine was assessed. Results: GLV-1h153 successfully facilitated serial long-term imaging of virotherapy, with PET signal intensity correlating to tumor response. GLV-1h153 colonization of tumors mediated radioiodine uptake at potentially therapeutic doses. Successful radiouptake required the presence of virus, adequate blood flow, and viable tissue, whereas loss of signal intensity was linked to tumor death and necrosis. Finally, combining systemically administered GLV-1h153 and 131I led to enhanced tumor kill when compared with virus or 131I alone (P < 0.01). Conclusion: GLV-1h153 is a promising oncolytic agent for the treatment, long-term imaging, and monitoring of therapeutic response in a xenograft model of pancreatic cancer. GLV-1h153 provided insight into tumor biologic activity and facilitated enhanced tumor kill when combined with systemic targeted radiotherapy. These results warrant further investigation into parameters and potential synergistic effects of combination therapy.


Surgery | 2013

Irreversible electroporation ablation of the liver can be detected with ultrasound B-mode and elastography.

Joyce T. Au; T. Peter Kingham; Kyonghwa Jun; Dana Haddad; Sepideh Gholami; Kelly Mojica; Sebastien Monette; Paula C. Ezell; Yuman Fong

BACKGROUND Irreversible electroporation (IRE) is a novel ablation technique that induces permanent membrane permeability and cell death. We are interested in ultrasound B-mode and elastography to monitor IRE ablation in the liver. METHODS Yorkshire pigs underwent IRE ablation of the liver and were imaged with ultrasound B-mode and elastography. Histologic evaluation of cell death by triphenyltetrazolium chloride and hematoxylin and eosin staining was performed. RESULTS Elastography showed that liver ablated by IRE exhibited increased tissue stiffness with a peak strain ratio of 2.22. The IRE lesion had a discrete border without bubble artifact, and the lesion size significantly correlated with area of cell death on histology. IRE ablation was unaffected by presence of large blood vessels or bile ducts. CONCLUSION IRE ablation led to increased tissue stiffness that was detectable by elastography and indicative of cell death. Elastography may complement B-mode ultrasonography to monitor IRE ablation of the liver.


Surgery | 2014

Oncolytic immunotherapy using recombinant vaccinia virus GLV-1h68 kills sorafenib-resistant hepatocellular carcinoma efficiently

Justin W. Ady; Jacqueline Heffner; Kelly Mojica; Clark Johnsen; Laurence J. Belin; Damon Love; Chin Tung Chen; Amudhan Pugalenthi; Elizabeth Klein; Nanhai G. Chen; Yong A. Yu; Aladar A. Szalay; Yuman Fong

BACKGROUND Sorafenib is the standard systemic therapy for unresectable or recurrent hepatocellular carcinoma (HCC) but adds minimal increase in survival. Therefore, there is a great need to develop novel therapies for advanced or recurrent HCC. One emerging field of cancer treatment involves oncolytic viruses that specifically infect, replicate within, and kill cancer cells. In this study, we examined the ability of GLV-1h68, a recombinant vaccinia virus derived from the vaccine strain that was used to eradicate smallpox, to kill sorafenib-resistant (SR) HCC cell lines. METHODS Four SR HCC cell lines were generated by repeated passage in the presence of sorafenib. Median inhibitory concentration was determined for all cell lines. The infectivity, viral replication, and cytotoxicity of GLV-1h68 were assayed for both parental and SR HCC cells. RESULTS Infectivity increased in a time and concentration-dependent manner in all cell lines. All cell lines supported efficient replication of virus. No difference between the rates of cell death between the parental and SR cell lines was observed. CONCLUSION Our results demonstrate that the oncolytic vaccinia virus GLV-1h68 kills both parental and SR HCC cell lines efficiently. This study indicates that patients who have failed treatment with sorafenib remain viable candidates for oncolytic therapy.


Molecular Therapy - Oncolytics | 2016

Tunneling nanotubes: an alternate route for propagation of the bystander effect following oncolytic viral infection

Justin W. Ady; Venugopal Thayanithy; Kelly Mojica; Phillip Y.-P. Wong; Joshua Carson; Prassanna Rao; Yuman Fong; Emil Lou

Tunneling nanotubes (TNTs) are ultrafine, filamentous actin-based cytoplasmic extensions which form spontaneously to connect cells at short and long-range distances. We have previously described long-range intercellular communication via TNTs connecting mesothelioma cells in vitro and demonstrated TNTs in intact tumors from patients with mesothelioma. Here, we investigate the ability of TNTs to mediate a viral thymidine kinase based bystander effect after oncolytic viral infection and administration of the nucleoside analog ganciclovir. Using confocal microscopy we assessed the ability of TNTs to propagate enhanced green fluorescent protein (eGFP), which is encoded by the herpes simplex virus NV1066, from infected to uninfected recipient cells. Using time-lapse imaging, we observed eGFP expressed in infected cells being transferred via TNTs to noninfected cells; additionally, increasing fluorescent activity in recipient cells indicated cell-to-cell transmission of the eGFP-expressing NV1066 virus had also occurred. TNTs mediated cell death as a form of direct cell-to-cell transfer following viral thymidine kinase mediated activation of ganciclovir, inducing a unique long-range form of the bystander effect through transmission of activated ganciclovir to nonvirus-infected cells. Thus, we provide proof-of-principle demonstration of a previously unknown and alternative mechanism for inducing apoptosis in noninfected recipient cells. The conceptual advance of this work is that TNTs can be harnessed for delivery of oncolytic viruses and of viral thymidine kinase activated drugs to amplify the bystander effect between cancer cells over long distances in stroma-rich tumor microenvironments.


Surgery | 2015

Oncolytic gene therapy with recombinant vaccinia strain GLV-2b372 efficiently kills hepatocellular carcinoma

Justin W. Ady; Clark Johnsen; Kelly Mojica; Jacqueline Heffner; Damon Love; Amudhan Pugalenthi; Laurence J. Belin; Nanhai G. Chen; Yong A. Yu; Aladar A. Szalay; Yuman Fong

BACKGROUND Hepatocellular carcinoma (HCC) commonly presents at a late stage when surgery is no longer a curative option. As such, novel therapies for advanced HCC are needed. Oncolytic viruses are a viable option for cancer therapy owing to their ability to specifically infect, replicate within, and kill cancer cells. In this study, we have investigated the ability of GLV-2b372, a novel light-emitting recombinant vaccinia virus derived from a wild-type Lister strain, to kill HCC. METHODS Four human HCC cell lines were assayed in vitro for infectivity and cytotoxicity. Viral replication was quantified via standard viral plaque assays. Flank HCC xenografts generated in athymic nude mice were treated with intratumoral GLV-2b372 to assess for tumor growth inhibition and viral biodistribution. RESULTS Infectivity occurred in a time- and concentration-dependent manner with 70% cell death in all cell lines by day 5. All cell lines supported efficient viral replication. At 25 days after infection, flank tumor volumes decreased by 50% whereas controls increased by 400%. Tumor tissue demonstrated substantial GLV-2b372 infection at 24 hours, 48 hours, and 2 weeks. CONCLUSION We demonstrate that GLV-2b372 efficiently kills human HCC in vitro and in vivo and is a viable treatment option for patients with HCC.


Cancer Research | 2017

miR-193b-regulated signaling networks serve as tumor suppressors in liposarcoma and promote adipogenesis in adipose-derived stem cells

Ying Zhang Mazzu; Yulan Hu; Rajesh Soni; Kelly Mojica; Li-Xuan Qin; Phaedra Agius; Zachary M. Waxman; Aleksandra Mihailovic; Nicholas D. Socci; Ronald C. Hendrickson; Thomas Tuschl; Samuel Singer

Well-differentiated and dedifferentiated liposarcomas (WDLS/DDLS) account for approximately 13% of all soft tissue sarcoma in adults and cause substantial morbidity or mortality in the majority of patients. In this study, we evaluated the functions of miRNA (miR-193b) in liposarcoma in vitro and in vivo Deep RNA sequencing on 93 WDLS, 145 DDLS, and 12 normal fat samples demonstrated that miR-193b was significantly underexpressed in DDLS compared with normal fat. Reintroduction of miR-193b induced apoptosis in liposarcoma cells and promoted adipogenesis in human adipose-derived stem cells (ASC). Integrative transcriptomic and proteomic analysis of miR-193b-target networks identified novel direct targets, including CRK-like proto-oncogene (CRKL) and focal adhesion kinase (FAK). miR-193b was found to regulate FAK-SRC-CRKL signaling through CRKL and FAK. miR-193b also stimulated reactive oxygen species signaling by targeting the antioxidant methionine sulfoxide reductase A to modulate liposarcoma cell survival and ASC differentiation state. Expression of miR-193b in liposarcoma cells was downregulated by promoter methylation, resulting at least in part from increased expression of the DNA methyltransferase DNMT1 in WDLS/DDLS. In vivo, miR-193b mimetics and FAK inhibitor (PF-562271) each inhibited liposarcoma xenograft growth. In summary, miR-193b not only functions as a tumor suppressor in liposarcoma but also promotes adipogenesis in ASC. Furthermore, this study reveals key tyrosine kinase and DNA methylation pathways in liposarcoma, some with immediate implications for therapeutic exploration. Cancer Res; 77(21); 5728-40. ©2017 AACR.


Surgery | 2015

Gene therapy using therapeutic and diagnostic recombinant oncolytic vaccinia virus GLV-1h153 for management of colorectal peritoneal carcinomatosis.

Clarisse Eveno; Kelly Mojica; Justin W. Ady; Daniel L. J. Thorek; Valerie A. Longo; Laurence J. Belin; Sepideh Gholami; Clark Johnsen; Pat Zanzonico; Nanhai Chen; Tony Yu; Aladar A. Szalay; Yuman Fong

BACKGROUND Peritoneal carcinomatosis (PC) is a terminal progression of colorectal cancer (CRC). Poor response to cytoreductive operation and chemotherapy coupled with the inability to reliably track disease progression by the use of established diagnostic methods, make this a deadly disease. We examined the effectiveness of the oncolytic vaccinia virus GLV-1h153 as a therapeutic and diagnostic vehicle. We believe that viral expression of the human sodium iodide transporter (hNIS) provides both real-time monitoring of viral therapy and effective treatment of colorectal peritoneal carcinomatosis (CRPC). METHODS Infectivity and cytotoxic effect of GLV-1h153 on CRC cell lines was assayed in vitro. Viral replication was examined by standard viral plaque assays. Orthotopic CRPC xenografts were generated in athymic nude mice and subsequently administered GLV-1h153 intraperitoneally. A decrease in tumor burden was assessed by mass. Orthotopic tumors were visualized by single-photon emission computed tomography/computed tomography after Iodine ((131)I) administration and by fluorescence optical imaging. RESULTS GLV-1h153 infected and killed CRC cells in a time- and concentration-dependent manner. Viral replication demonstrated greater than a 2.35 log increase in titer over 4 days. Intraperitoneal treatment of orthotopic CRPC xenografts resulted in a substantial decrease in tumor burden. Infection of orthotopic xenografts was therapeutic and facilitated monitoring by (131)I-single-photon emission computed tomography/computed tomography via expression of hNIS in infected tissue. CONCLUSION GLV-1h153 kills CRC in vitro effectively and decreases tumor burden in vivo. We demonstrate that GLV-1h153 can be used as an agent to provide accurate delineation of tumor burden in vivo. These findings indicate that GLV-1h153 has potential for use as a therapeutic and diagnostic agent in the treatment of CRPC.


Cancer Gene Therapy | 2015

Recombinant vaccinia virus GLV-1h68 is a promising oncolytic vector in the treatment of cholangiocarcinoma

Amudhan Pugalenthi; Kelly Mojica; Justin W. Ady; Clark Johnsen; Damon Love; Nanhai G. Chen; Richard J. Aguilar; Aladar A. Szalay; Yuman Fong

Although early stage cholangiocarcinoma (CC) can be cured by surgical extirpation, the options for treatment of advanced stage CC are very few and suboptimal. Oncolytic virotherapy using replication-competent vaccinia virus (VACV) is a promising new strategy to treat human cancers. The ability of oncolytic VACV GLV-1h68 to infect, replicate in, and lyse three human CC cell lines was assayed in vitro and in subcutaneous flank xenografts in athymic nude mice. In this study, we have demonstrated that GLV-1h68 effectively infects and lyses three CC cell lines (KMC-1, KMBC, and KMCH-1) in vitro. Expression of the viral marker gene ruc-gfp facilitated real-time monitoring of infection and replication. Furthermore in athymic nude mice, a single dose of GLV-1h68 significantly suppressed tumor growth. The treatment was well tolerated in all animals. Recombinant VACV GLV-1h68 has significant oncolytic ability against CC both in vitro and in vivo. GLV-1h68 has the potential to be used clinically as a therapeutic agent against CC.


Cancer Research | 2016

Abstract 929: Tunneling nanotube conduits facilitate the bystander effect after oncolytic viral infection

Emil Lou; Justin W. Ady; Venugopal Thayanithy; Kelly Mojica; Joshua Carson; Prassanna Rao; Yuman Fong

Background: Oncolytic viruses have come to the forefront of cancer therapeutics following FDA approval for treatment of metastatic melanoma in 2015. Efficacy of viral therapy is enhanced by the bystander effect, a cellular phenomenon that amplifies the effects of the virus following activation of the prodrug ganciclovir (GCV) by virally-expressed thymidine kinase (TK) and intercellular spread of GCV via gap junctions. In the complex and stroma-rich tumor microenvironment, gap junctions may not completely account for cell-to-cell communication. Tunneling nanotubes (TNTs) are a novel and recently characterized alternative form of direct cell-to-cell communication in the tumor matrix. TNTs are fine, long, F actin-based cell extensions that serve as short and long-range conduits for efficient transfer of cellular cargo. Here, we investigate the ability of TNTs to mediate a TK-based bystander effect after oncolytic viral infection and administration of GCV. Methods: We infected 3 mesothelioma cell lines with NV1066, a mutant replication-competent strain of herpes simplex virus-1 (HSV-1) that encodes viral eGFP and viral TK. Confocal microscopy and time-lapse imaging were performed 12-36 hours later. A modified Transwell assay was used to separate infected cells from uninfected cells to assess TNT propagation of eGFP-tagged NV1066. GCV was added to infected cells in the top chamber to assess TNT propagation of TK-activated GCV. Apoptosis was measured using TUNEL assay in the bottom chamber to quantify the extent of the bystander effect. Results: Confocal microscopy demonstrated effective intercellular transfer of GFP-tagged virus between cells via TNTs prior to oncolysis. Quantification of TUNEL-positive cells at 48 hours indicated that addition of NV1066 to the top chamber resulted in 33% of the initially uninfected cells in the bottom chamber dying by 48 hours. The addition of GCV to virally infected cells in the top chamber significantly increased apoptosis in recipient cells in the bottom chamber, from 33% to 71%, producing a 2.3-fold increase in cell killing attributed to TNT transfer of viral TK-activated GCV (p = 0.007). Thus, TNTs were shown to transfer viral TK-activated GCV to non-infected cells, leading to cell death via a long-range form of the bystander effect. Conclusions: Here we demonstrate that TNTs provide a previously unknown and alternative mechanism for the bystander effect in which viral TK-activated GCV is transferred via TNTs and induces apoptosis in non-infected recipient cells. Citation Format: Emil Lou, Justin Ady, Venugopal Thayanithy, Kelly Mojica, Joshua Carson, Prassanna Rao, Yuman Fong. Tunneling nanotube conduits facilitate the bystander effect after oncolytic viral infection. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 929.


Cancer Research | 2012

Abstract 5660: Vaccinia virus GLV-1h153 in combination with 131I-iodine shows increased efficiency in treating triple-negative breast cancer in vivo

Sepideh Gholami; Chun-Hao Chen; Emil Lou; Andrew A. Marano; Kelly Mojica; Nanhai G. Chen; Richard J. Aguilar; Laurence J. Belin; Valerie A. Longo; Pat Zanzonico; Aladar A. Szalay; Yuman Fong

Proceedings: AACR 103rd Annual Meeting 2012‐‐ Mar 31‐Apr 4, 2012; Chicago, IL BACKGROUND: Triple-negative breast cancers (TNBC) are aggressive tumors due to their inherent biology and the lack of receptors for hormonal/targeted therapy, namely estrogen, progesterone, and Her2/neu. The human sodium iodide symporter (hNIS) is a naturally occurring protein in some human breast and thyroid tissue which enables cells to concentrate iodine, including radioiodide. In this study, we investigated the therapeutic efficacy of a new oncolytic vaccinia virus, GLV-1h153 carrying the hNIS, in combination with radioiodine in a TNBC murine model. METHODS: GLV-1h153, a replication-competent vaccinia virus, was tested against the TNBC cell lines MDA-MB-231, MDA-MB-468, HCC-1937, and HCC-1143 at multiplicities of infections (MOI) of 0.1, 1.0, and 5. Cytotoxicity and viral replication were determined. Mammary fat pad tumors were generated in athymic nude mice with MDA-MB-231 cells. A subset of xenografts were infected with GLV-1h153 and ∼150 μCi of 124I-iodine was administered. Serial Focus 120 microPET were obtained for 131I dosimetry calculations. For the combination therapy study, 14 days after cell implantation, xenografts were treated with intratumoral injection of GLV-1h153 or PBS. One week after viral injection (day 21), xenografts were further randomized into 4 treatments groups: GLV-1h153 alone, GLV-1h153 and iodine (∼5 mCi of 131I), iodine alone, or PBS and followed for tumor growth. RESULTS: Greater than 90% cell kill was achieved in all cell lines within 5 days at an MOI of 5.0. GLV-1h153 replicated efficiently in all cell lines with a peak titer of 2.6 x107 viral plaque forming units per ml (>1300-fold increase from the initial viral dose) by day 4 in cell line MDA-MB-468. Only infected tumors were identified via PET scanning compared to controls. In vivo, administration of systemic radioiodine in combination with GLV-1h153 resulted in greater tumor regression, 24 mm3 compared to 146 mm3 for the viral-treated group only (p<0.05; days 21-40), a six-fold difference. CONCLUSION: GLV-1h153 infected, replicated in, and killed all TNBC cell lines effectively. This study is the first to our knowledege to demonstrate killing of TNBC by a novel vaccinia virus in combination with radioactive 131I-iodine in an in vivo xenograft model. Our results suggest that GLV-1h153 is a promising therapeutic agent in combination with 131I and merits further testing in the clinical setting. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 5660. doi:1538-7445.AM2012-5660

Collaboration


Dive into the Kelly Mojica's collaboration.

Top Co-Authors

Avatar

Yuman Fong

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Justin W. Ady

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Laurence J. Belin

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Pat Zanzonico

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Sepideh Gholami

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Valerie A. Longo

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Clark Johnsen

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Amudhan Pugalenthi

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Damon Love

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge