Anne Moon
University of California, Berkeley
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Anne Moon.
Nature Medicine | 2013
Jeong Heo; Tony Reid; Leyo Ruo; Caroline J. Breitbach; Steven C. Rose; Mark Bloomston; Mong Cho; Ho Yeong Lim; Hyun Cheol Chung; Chang Won Kim; James R. Burke; Riccardo Lencioni; Theresa Hickman; Anne Moon; Yeon Sook Lee; Mi Kyeong Kim; Manijeh Daneshmand; Kara S DuBois; Lara Longpre; Minhtran Ngo; Cliona M. Rooney; John C. Bell; Byung Geon Rhee; Richard H. Patt; Tae Ho Hwang; David Kirn
Oncolytic viruses and active immunotherapeutics have complementary mechanisms of action (MOA) that are both self amplifying in tumors, yet the impact of dose on subject outcome is unclear. JX-594 (Pexa-Vec) is an oncolytic and immunotherapeutic vaccinia virus. To determine the optimal JX-594 dose in subjects with advanced hepatocellular carcinoma (HCC), we conducted a randomized phase 2 dose-finding trial (n = 30). Radiologists infused low- or high-dose JX-594 into liver tumors (days 1, 15 and 29); infusions resulted in acute detectable intravascular JX-594 genomes. Objective intrahepatic Modified Response Evaluation Criteria in Solid Tumors (mRECIST) (15%) and Choi (62%) response rates and intrahepatic disease control (50%) were equivalent in injected and distant noninjected tumors at both doses. JX-594 replication and granulocyte-macrophage colony-stimulating factor (GM-CSF) expression preceded the induction of anticancer immunity. In contrast to tumor response rate and immune endpoints, subject survival duration was significantly related to dose (median survival of 14.1 months compared to 6.7 months on the high and low dose, respectively; hazard ratio 0.39; P = 0.020). JX-594 demonstrated oncolytic and immunotherapy MOA, tumor responses and dose-related survival in individuals with HCC.
Nature | 2011
Caroline J. Breitbach; James Burke; Derek J. Jonker; Joe Stephenson; Andrew R. Haas; Laura Quan Man Chow; Jorge Nieva; Tae Ho Hwang; Anne Moon; Richard H. Patt; Adina Pelusio; Fabrice Le Boeuf; Joseph K. Burns; Laura Evgin; Naomi De Silva; Sara Cvancic; Terri Robertson; Ji Eun Je; Yeon Sook Lee; Kelley Parato; Jean-Simon Diallo; Aaron Fenster; Manijeh Daneshmand; John C. Bell; David Kirn
The efficacy and safety of biological molecules in cancer therapy, such as peptides and small interfering RNAs (siRNAs), could be markedly increased if high concentrations could be achieved and amplified selectively in tumour tissues versus normal tissues after intravenous administration. This has not been achievable so far in humans. We hypothesized that a poxvirus, which evolved for blood-borne systemic spread in mammals, could be engineered for cancer-selective replication and used as a vehicle for the intravenous delivery and expression of transgenes in tumours. JX-594 is an oncolytic poxvirus engineered for replication, transgene expression and amplification in cancer cells harbouring activation of the epidermal growth factor receptor (EGFR)/Ras pathway, followed by cell lysis and anticancer immunity. Here we show in a clinical trial that JX-594 selectively infects, replicates and expresses transgene products in cancer tissue after intravenous infusion, in a dose-related fashion. Normal tissues were not affected clinically. This platform technology opens up the possibility of multifunctional products that selectively express high concentrations of several complementary therapeutic and imaging molecules in metastatic solid tumours in humans.
Lancet Oncology | 2008
Byeong Ho Park; Tae-Ho Hwang; Ta-Chiang Liu; Daniel Y. Sze; Jae Seok Kim; Hyuk Chan Kwon; Sung Yong Oh; Sang Young Han; Jin Han Yoon; Sook Hee Hong; Anne Moon; Kelly Speth; Chohee Park; Young Joo Ahn; Manijeh Daneshmand; Byung Geon Rhee; Herbert M. Pinedo; John C. Bell; David Kirn
BACKGROUND JX-594 is a targeted oncolytic poxvirus designed to selectively replicate in and destroy cancer cells with cell-cycle abnormalities and epidermal growth factor receptor (EGFR)-ras pathway activation. Direct oncolysis plus granulocyte-macrophage colony-stimulating factor (GM-CSF) expression also stimulates shutdown of tumour vasculature and antitumoral immunity. We aimed to assess intratumoral injection of JX-594 in patients with refractory primary or metastatic liver cancer. METHODS Between Jan 4, 2006, and July 4, 2007, 14 patients with histologically confirmed refractory primary or metastatic liver tumours (up to 10.9 cm total diameter) that were amenable to image-guided intratumoral injections were enrolled into this non-comparative, open-label, phase I dose-escalation trial (standard 3x3 design; two to six patients for each dose with 12-18 estimated total patients). Patients received one of four doses of intratumoral JX-594 (10(8) plaque-forming units [pfu], 3x10(8) pfu, 10(9) pfu, or 3x10(9) pfu) every 3 weeks at Dong-A University Hospital (Busan, South Korea). Patients were monitored after treatment for at least 48 h in hospital and for at least 4 weeks as out-patients. Adverse event-monitoring according to the National Cancer Institute Common Toxicity Criteria (version 3) and standard laboratory toxicity grading for haematology, liver and renal function, coagulation studies, serum chemistry, and urinalysis were done. The primary aims were to ascertain the maximum-tolerated dose (MTD) and safety of JX-594 treatment. Data were also collected on pharmacokinetics, pharmacodynamics, and efficacy. Analysis was per protocol. This study is registered with ClinicalTrials.gov, number NCT00629759. FINDINGS Of 22 patients with liver tumours who were assessed for eligibility, eight patients did not meet inclusion criteria. Therefore, 14 patients, including those with hepatocellular, colorectal, melanoma, and lung cancer, were enrolled. Patients were heavily pretreated (5.6 previous treatments, SD 2.8, range 2.0-12.0) and had large tumours (7.0 cm diameter, SD 2.7, range 1.8-10.9). Patients received a mean of 3.4 (SD 2.2, range 1.0-8.0) cycles of JX-594. All patients were evaluable for toxicity. All patients experienced grade I-III flu-like symptoms, and four had transient grade I-III dose-related thrombocytopenia. Grade III hyperbilirubinaemia was dose-limiting in both patients at the highest dose; the MTD was therefore 1x10(9) pfu. JX-594 replication-dependent dissemination in blood was shown, with resultant infection of non-injected tumour sites. GM-CSF expression resulted in grade I-III increases in neutrophil counts in four of six patients at the MTD. Tumour responses were shown in injected and non-injected tumours. Ten patients were radiographically evaluable for objective responses; non-evaluable patients had contraindications to contrast medium (n=2) or no post-treatment scans (n=2). According to Response Evaluation Criteria in Solid Tumors (RECIST), three patients had partial response, six had stable disease, and one had progressive disease. INTERPRETATION Intratumoral injection of JX-594 into primary or metastatic liver tumours was generally well-tolerated. Direct hyperbilirubinaemia was the dose-limiting toxicity. Safety was acceptable in the context of JX-594 replication, GM-CSF expression, systemic dissemination, and JX-594 had anti-tumoral effects against several refractory carcinomas. Phase II trials are now underway.
Molecular Therapy | 2011
Tae-Ho Hwang; Anne Moon; James Burke; Antoni Ribas; Joe Stephenson; Caroline J. Breitbach; Manijeh Daneshmand; Naomi De Silva; Kelley Parato; Jean-Simon Diallo; Yeon-Sook Lee; Ta-Chiang Liu; John C. Bell; David Kirn
JX-594 is a targeted and granulocyte macrophage-colony stimulating factor (GM-CSF)-expressing oncolytic poxvirus designed to selectively replicate in and destroy cancer cells through viral oncolysis and tumor-specific immunity. In order to study the mechanisms-of-action (MOA) of JX-594 in humans, a mechanistic proof-of-concept clinical trial was performed at a low dose equivalent to ≤10% of the maximum-tolerated dose (MTD) in other clinical trials. Ten patients with previously treated stage IV melanoma were enrolled. Tumors were injected weekly for up to nine total treatments. Blood samples and tumor biopsies were analyzed for evidence of transgene activity, virus replication, and immune stimulation. The β-galactosidase (β-gal) transgene was expressed in all patients as evidenced by antibody induction. Six patients had significant induction of GM-CSF-responsive white blood cell (WBC) subsets such as neutrophils (25-300% increase). JX-594 replication and subsequent shedding into blood was detectable in five patients after cycles 1-9. Tumor biopsies demonstrated JX-594 replication, perivascular lymphocytic infiltration, and diffuse tumor necrosis. Mild flu-like symptoms were the most common adverse events. In sum, JX-594 replication, oncolysis, and expression of both transgenes were demonstrated; replication was still evident after multiple cycles. These findings have implications for further clinical development of JX-594 and other transgene-armed oncolytic viruses.
Cancer Research | 2013
Caroline J. Breitbach; Rozanne Arulanandam; Naomi De Silva; Steve H. Thorne; Richard H. Patt; Manijeh Daneshmand; Anne Moon; Carolina S. Ilkow; James R. Burke; Tae-Ho Hwang; Jeong Heo; Mong Cho; Hannah Chen; Fernando A. Angarita; Christina L. Addison; J. Andrea McCart; John C. Bell; David Kirn
Efforts to selectively target and disrupt established tumor vasculature have largely failed to date. We hypothesized that a vaccinia virus engineered to target cells with activation of the ras/MAPK signaling pathway (JX-594) could specifically infect and express transgenes (hGM-CSF, β-galactosidase) in tumor-associated vascular endothelial cells in humans. Efficient replication and transgene expression in normal human endothelial cells in vitro required either VEGF or FGF-2 stimulation. Intravenous infusion in mice resulted in virus replication in tumor-associated endothelial cells, disruption of tumor blood flow, and hypoxia within 48 hours; massive tumor necrosis ensued within 5 days. Normal vessels were not affected. In patients treated with intravenous JX-594 in a phase I clinical trial, we showed dose-dependent endothelial cell infection and transgene expression in tumor biopsies of diverse histologies. Finally, patients with advanced hepatocellular carcinoma, a hypervascular and VEGF-rich tumor type, were treated with JX-594 on phase II clinical trials. JX-594 treatment caused disruption of tumor perfusion as early as 5 days in both VEGF receptor inhibitor-naïve and -refractory patients. Toxicities to normal blood vessels or to wound healing were not evident clinically or on MRI scans. This platform technology opens up the possibility of multifunctional engineered vaccinia products that selectively target and infect tumor-associated endothelial cells, as well as cancer cells, resulting in transgene expression, vasculature disruption, and tumor destruction in humans systemically.
Molecular Therapy | 2011
Jeong Heo; Caroline J. Breitbach; Anne Moon; Chang Won Kim; Rick Patt; Mi Kyung Kim; Yu Kyung Lee; Sung Yong Oh; Hyun Young Woo; Kelley Parato; Julia Rintoul; Theresa Falls; Theresa Hickman; Byung-Geon Rhee; John C. Bell; David Kirn; Tae-Ho Hwang
JX-594 is a targeted and granulocyte-macrophage colony stimulating factor (GM-CSF) expressing oncolytic poxvirus designed to selectively replicate in and destroy cancer cells through viral oncolysis and tumor-specific immunity. In a phase 1 trial, JX-594 injection into hepatocellular carcinoma (HCC) was well-tolerated and associated with viral replication, decreased tumor perfusion, and tumor necrosis. We hypothesized that JX-594 and sorafenib, a small molecule inhibitor of B-raf and vascular endothelial growth factor receptor (VEGFR) approved for HCC, would have clinical benefit in combination given their demonstrated efficacy in HCC patients and their complementary mechanisms-of-action. HCC cell lines were uniformly sensitive to JX-594. Anti-raf kinase effects of concurrent sorafenib inhibited JX-594 replication in vitro, whereas sequential therapy was superior to either agent alone in murine tumor models. We therefore explored pilot safety and efficacy of JX-594 followed by sorafenib in three HCC patients. In all three patients, sequential treatment was (i) well-tolerated, (ii) associated with significantly decreased tumor perfusion, and (iii) associated with objective tumor responses (Choi criteria; up to 100% necrosis). HCC historical control patients on sorafenib alone at the same institutions had no objective tumor responses (0 of 15). Treatment of HCC with JX-594 followed by sorafenib has antitumoral activity, and JX-594 may sensitize tumors to subsequent therapy with VEGF/VEGFR inhibitors.
Science Translational Medicine | 2013
Mi Kyung Kim; Caroline J. Breitbach; Anne Moon; Jeong Heo; Yu Kyoung Lee; Mong Cho; Jun Woo Lee; Seong-Geun Kim; Dae Hwan Kang; John C. Bell; Byeong Ho Park; David Kirn; Tae-Ho Hwang
The oncolytic and immunotherapeutic vaccinia virus Pexa-Vec (JX-594) induces antibody-mediated complement-dependent cancer cell cytolysis in rabbits and cancer patients, prolonging their survival. The Enemy of My Enemy Is My Friend: Virotherapy for Cancer Oncolytic viruses, which can kill cancer cells directly and by stimulating the patient’s immune system, have some clear advantages over other immune approaches to cancer treatment. They do not induce autoimmunity, they do not have to be custom-made for each patient, and they are not specific to tumors that express a particular antigen. Previous works with these viruses have shown promising results, and some of them are now in clinical trials. However, the steps by which these viruses exert their effects against the cancer cells in animals and humans are not yet fully understood. Now, the work by Kim et al. uncovers some of the mechanism for the action of Pexa-Vec, a vaccinia virus–derived oncolytic vaccine, which is already being tested in patients with multiple types of advanced cancers. In a rabbit model of squamous cell carcinoma, the authors demonstrated the induction of antibody-mediated complement-dependent cytotoxicity (CDC) after treatment with Pexa-Vec. The resulting antitumor activity could be transferred from one rabbit to another by transferring serum, even long after the virus was administered and then cleared from the blood, providing additional evidence for the role of antibodies. Serum from human patients treated with Pexa-Vec also induced CDC against cancer cells, primarily against cancer of the same type. For example, serum from a renal cancer patient treated with the virus was most effective at triggering CDC against renal cancer cells, consistent with a specific antibody-mediated process. The ability of each patient’s serum to induce CDC against cultured cancer cells correlated with that patient’s survival. Larger trials in human patients will be required to confirm the pilot study results presented here and build on the current understanding of oncolytic virotherapy and the role of CDC. In addition, further studies will need to investigate the role of other immune mechanisms, such as cell-mediated immunity, in the effects of oncolytic virus vaccines. In the meantime, the current study highlights the role of Pexa-Vec in the induction of antitumor CDC and shows its effects on survival, even in patients with late-stage cancer who have few other options. Oncolytic viruses cause direct cytolysis and cancer-specific immunity in preclinical models. The goal of this study was to demonstrate induction of functional anticancer immunity that can lyse target cancer cells in humans. Pexa-Vec (pexastimogene devacirepvec; JX-594) is a targeted oncolytic and immunotherapeutic vaccinia virus engineered to express human granulocyte-macrophage colony-stimulating factor (GM-CSF). Pexa-Vec demonstrated replication, GM-CSF expression, and tumor responses in previous phase 1 trials. We now evaluated whether Pexa-Vec induced functional anticancer immunity both in the rabbit VX2 tumor model and in patients with diverse solid tumor types in phase 1. Antibody-mediated complement-dependent cancer cell cytotoxicity (CDC) was induced by intravenous Pexa-Vec in rabbits; transfer of serum from Pexa-Vec–treated animals to tumor-bearing animals resulted in tumor necrosis and improved survival. In patients with diverse tumor types treated on a phase 1 trial, CDC developed within 4 to 8 weeks in most patients; normal cells were resistant to the cytotoxic effects. T lymphocyte activation in patients was evidenced by antibody class switching. We determined that patients with the longest survival duration had the highest CDC activity, and identified candidate target tumor cell antigens. Thus, we demonstrated that Pexa-Vec induced polyclonal antibody–mediated CDC against multiple tumor antigens both in rabbits and in patients with diverse solid tumor types.
Molecular Therapy | 2015
Timothy P. Cripe; Minhtran Ngo; James I. Geller; Chrystal U. Louis; Mark A. Currier; John M. Racadio; Alexander J. Towbin; Cliona M. Rooney; Adina Pelusio; Anne Moon; Tae-Ho Hwang; James Burke; John C. Bell; David Kirn; Caroline J. Breitbach
Pexa-Vec (pexastimogene devacirepvec, JX-594) is an oncolytic and immunotherapeutic vaccinia virus designed to destroy cancer cells through viral lysis and induction of granulocyte-macrophage colony-stimulating factor (GM-CSF)-driven tumor-specific immunity. Pexa-Vec has undergone phase 1 and 2 testing alone and in combination with other therapies in adult patients, via both intratumoral and intravenous administration routes. We sought to determine the safety of intratumoral administration in pediatric patients. In a dose-escalation study using either 10(6) or 10(7) plaque-forming units per kilogram, we performed one-time injections in up to three tumor sites in five pediatric patients and two injections in one patient. Ages at study entry ranged from 4 to 21 years, and their cancer diagnoses included neuroblastoma, hepatocellular carcinoma, and Ewing sarcoma. All toxicities were ≤ grade 3. The most common side effects were sinus fever and sinus tachycardia. All three patients at the higher dose developed asymptomatic grade 1 treatment-related skin pustules that resolved within 3-4 weeks. One patient showed imaging evidence suggestive of antitumor biological activity. The two patients tested for cellular immunoreactivity to vaccinia antigens showed strong responses. Overall, our study suggests Pexa-Vec is safe to administer to pediatric patients by intratumoral administration and could be studied further in this patient population.
Molecular Therapy | 2015
Se Hoon Park; Caroline J. Breitbach; Jeeyun Lee; Joon Oh Park; Ho Yeong Lim; Won Ki Kang; Anne Moon; Jae-Hee Mun; Erica M Sommermann; Liliana Maruri Avidal; Rick Patt; Adina Pelusio; James R. Burke; Tae-Ho Hwang; David Kirn; Young Suk Park
Fifteen patients with treatment-refractory colorectal cancer were enrolled on a phase 1b study of Pexa-Vec (pexastimogene devacirepvec; JX-594), an oncolytic and immunotherapeutic vaccinia designed to selectively replicate in cancer cells. Pexa-Vec was administered intravenously every 14 days, at dose levels of 1 × 10(6), 1 × 10(7), or 3 × 10(7) plaque-forming units (pfu)/kg. The primary endpoint was to determine the maximum tolerated dose. Secondary endpoints were pharmacokinetics and pharmacodynamics as well as antitumor activity. Patients were heavily pretreated (mean 4.5 lines of therapy). All patients received at least two Pexa-Vec doses (median = 4; range = 2-4). No dose-limiting toxicities were reported, and the maximum tolerated dose was not reached. The most common adverse events were grade 1/2 flu-like symptoms, generally lasting <24 hours. During the first and last cycles, genome pharmacokinetics were unchanged. Infectious pfu could be detected in plasma up to 2 hours after cycle 1 and up to 30 minutes after cycle 4 (when antivaccinia antibody titers are known to have peaked). Ten patients (67%) had radiographically stable disease. Given the acceptable safety profile of multiple intravenous Pexa-Vec infusions in patients with treatment-refractory colorectal cancer, further trials evaluating efficacy of intravenous Pexa-Vec, as monotherapy or in combination with chemotherapeutic agents, is warranted in this patient population.
Molecular Therapy | 2015
Herbert J. Zeh; Stephanie Downs-Canner; J. Andrea McCart; Zong Sheng Guo; Uma N. M. Rao; Lekshmi Ramalingam; Stephen H. Thorne; Heather L. Jones; Pawel Kalinski; Eva Wieckowski; Mark E. O'Malley; Manijeh Daneshmand; Kang Hu; John C. Bell; Tae-Ho Hwang; Anne Moon; Caroline J. Breitbach; David Kirn; David L. Bartlett
Oncolytic viral therapy utilizes a tumor-selective replicating virus which preferentially infects and destroys cancer cells and triggers antitumor immunity. The Western Reserve strain of vaccinia virus (VV) is the most virulent strain of VV in animal models and has been engineered for tumor selectivity through two targeted gene deletions (vvDD). We performed the first-in-human phase 1, intratumoral dose escalation clinical trial of vvDD in 16 patients with advanced solid tumors. In addition to safety, we evaluated signs of vvDD replication and spread to distant tumors, pharmacokinetics and pharmacodynamics, clinical and immune responses to vvDD. Dose escalation proceeded without dose-limiting toxicities to a maximum feasible dose of 3 × 10(9) pfu. vvDD replication in tumors was reproducible. vvDD genomes and/or infectious particles were recovered from injected (n = 5 patients) and noninjected (n = 2 patients) tumors. At the two highest doses, vvDD genomes were detected acutely in blood in all patients while delayed re-emergence of vvDD genomes in blood was detected in two patients. Fifteen of 16 patients exhibited late symptoms, consistent with ongoing vvDD replication. In summary, intratumoral injection of the oncolytic vaccinia vvDD was well-tolerated in patients and resulted in selective infection of injected and noninjected tumors and antitumor activity.