Network


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

Hotspot


Dive into the research topics where Christopher P. Leamon is active.

Publication


Featured researches published by Christopher P. Leamon.


Drug Discovery Today | 2001

Folate-mediated targeting : from diagnostics to drug and gene delivery

Christopher P. Leamon; Philip S. Low

The covalent attachment of the vitamin folic acid to almost any molecule yields a conjugate that can be endocytosed into folate receptor-bearing cells. Because folate receptors are significantly overexpressed in the majority of human cancers, this methodology is currently being investigated for the selective delivery of imaging and therapeutic agents to tumor tissue. Phase I and II clinical studies for the first folate-containing imaging agent were initiated in 1999, and clinical trials of folate-targeted therapeutic agents should soon follow. This review will summarize folate-mediated drug delivery and highlight those techniques undergoing active preclinical or clinical investigation.


Journal of Clinical Oncology | 2010

PRECEDENT: A Randomized Phase II Trial Comparing Vintafolide (EC145) and Pegylated Liposomal Doxorubicin (PLD) in Combination Versus PLD Alone in Patients With Platinum-Resistant Ovarian Cancer

R. Wendel Naumann; Robert L. Coleman; Robert A. Burger; Edward A. Sausville; Elzbieta Kutarska; Sharad A. Ghamande; Nashat Y. Gabrail; Stephen E. DePasquale; Elżbieta Nowara; Lucy Gilbert; Robert H. Gersh; Michael Teneriello; Wael A. Harb; Panagiotis A. Konstantinopoulos; Richard T. Penson; James Symanowski; Chandra D. Lovejoy; Christopher P. Leamon; David Morgenstern; Richard A. Messmann

PURPOSE Vintafolide (EC145) is a folic acid-desacetylvinblastine conjugate that binds to the folate receptor (FR), which is expressed on the majority of epithelial ovarian cancers. This randomized phase II trial evaluated vintafolide combined with pegylated liposomal doxorubicin (PLD) compared with PLD alone. The utility of an FR-targeted imaging agent, (99m)Tc-etarfolatide (EC20), in selecting patients likely to benefit from vintafolide was also examined. PATIENTS AND METHODS Women with recurrent platinum-resistant ovarian cancer who had undergone ≤ two prior cytotoxic regimens were randomly assigned at a 2:1 ratio to PLD (50 mg/m(2) intravenously [IV] once every 28 days) with or without vintafolide (2.5 mg IV three times per week during weeks 1 and 3). Etarfolatide scanning was optional. The primary objective was to compare progression-free survival (PFS) between the groups. RESULTS The intent-to-treat population comprised 149 patients. Median PFS was 5.0 and 2.7 months for the vintafolide plus PLD and PLD-alone arms, respectively (hazard ratio [HR], 0.63; 95% CI, 0.41 to 0.96; P = .031). The greatest benefit was observed in patients with 100% of lesions positive for FR, with median PFS of 5.5 compared with 1.5 months for PLD alone (HR, 0.38; 95% CI, 0.17 to 0.85; P = .013). The group of patients with FR-positive disease (10% to 90%) experienced some PFS improvement (HR, 0.873), whereas patients with disease that did not express FR experienced no PFS benefit (HR, 1.806). CONCLUSION Vintafolide plus PLD is the first combination to demonstrate an improvement over standard therapy in a randomized trial of patients with platinum-resistant ovarian cancer. Etarfolatide can identify patients likely to benefit from vintafolide.


Cancer Research | 2007

Preclinical Evaluation of EC145, a Folate-Vinca Alkaloid Conjugate

Joseph A. Reddy; Ryan Dorton; Elaine Westrick; Alicia Dawson; Terri Smith; Le-Cun Xu; Marilynn Vetzel; Paul J. Kleindl; Iontcho Radoslavov Vlahov; Christopher P. Leamon

We recently developed a new group of folate-conjugated Vinca alkaloids, one of which, EC145, emerged as a candidate for clinical development. Brief treatment of nude mice bearing approximately 100 mm(3) folate receptor-positive human xenografts led to complete response (CR) in 5/5 mice and cures (i.e., remission without a relapse for >90 days post-tumor implantation) in 4/5 mice. Multiple CRs and cures were also noted when EC145 was used to treat mice initially bearing tumors as large as 750 mm(3). Likewise, complete cures (5/5) resulted following the treatment of an aggressive folate receptor-positive J6456 lymphoma model. The activity of EC145 was not accompanied by noticeable weight loss or major organ tissue degeneration. Furthermore, no significant antitumor activity (0/5 CR) was observed in EC145-treated animals that were co-dosed with an excess of a benign folate ligand, thus demonstrating the target-specific activity of EC145. The enhanced therapeutic index due to folate conjugation was also evidenced by the fact that the unconjugated drug (desacetylvinblastine monohydrazide) was found to be completely inactive when administered at nontoxic dose levels and only marginally active when given at highly toxic dose levels. Subsequent dose regimen studies confirmed that EC145 given on a more frequent, qdx5 schedule resulted in the most effective antitumor response as compared with an equivalent total dose given on thrice- or single-injection-per-week schedule. Taken together, these studies show that EC145 has significant antiproliferative activity and tolerability, thus lending support to an ongoing phase 1 trial for the treatment of advanced malignancies.


Bioconjugate Chemistry | 2012

Engineering Folate–Drug Conjugates to Target Cancer: From Chemistry to Clinic

Iontcho Radoslavov Vlahov; Christopher P. Leamon

The folate receptor (FR) is a potentially useful biological target for the management of many human cancers. This membrane protein binds extracellular folates with very high affinity and, through an endocytic process, physically delivers them inside the cell for biological consumption. There are now many examples of how this physiological system can be exploited for the targeted delivery of biologically active molecules to cancer. In fact, strong preclinical as well as emerging clinical evidence exists showing how FR-positive cancers can be (i) anatomically identified using folate conjugates of radiodiagnostic imaging agents and (ii) effectively treated with companion folate-targeted chemotherapies. While the biological results are compelling, it is of equal importance to understand the conjugation chemistries that were developed to produce these active molecules. Therefore, this review will focus on the methods utilized to construct folate-based small-molecule drug conjugates (SMDCs), with particular attention focused on modular design, hydrophilic spacers, and self-immolative linkers.


Journal of Drug Targeting | 1994

Selective Targeting of Malignant Cells with Cytotoxin-Folate Conjugates

Christopher P. Leamon; Philip S. Low

Previous work has shown that proteins can be nondestructively delivered into the cytoplasm of folate receptor-bearing cells if the proteins are conjugated to folic acid prior to addition to cells. In view of other reports suggesting the membrane receptor for folic acid is vastly overexpressed on tumor cells, we decided to explore whether malignant cells might be selectively targeted in a co-culture with nontransformed cells using ligated folate as the targeting agent. Exploiting the cytotoxicity of the ribosome-inactivating toxin, momordin, to assay successful intracellular protein delivery, we demonstrate that HeLa and KB cells (two malignant human cell lines) can be selectively and quantitatively killed in co-cultures with WI38 and Hs67 cells (two normal human cell types). Not only are the normal cells not damaged by treatment of the co-cultures with momordin-folate, but their rates of proliferation actually accelerate, presumably due to increased availability of nutrients otherwise consumed by the transformed cells. Analysis of folate receptor number before and after momordin-folate treatment indicates that receptor density may be a good predictor of toxin-folate sensitivity. Taken together, the data suggest that conjugates of folic acid with cytotoxic proteins warrant further examination as possible tumor-specific chemotherapeutic agents.


Drug Resistance Updates | 2014

The folate receptor as a rational therapeutic target for personalized cancer treatment

Yehuda G. Assaraf; Christopher P. Leamon; Joseph A. Reddy

Conventional cancer treatment modalities have several limitations including lack of sufficient efficacy, serious untoward toxicity, as well as innate and acquired drug resistance. In contrast, targeted imaging agents can identify patients with receptors overexpressed on the surface of cancer cells, thus allowing appropriate selection of patients for personalized treatment with a desirable targeted therapeutic. The folate receptor (FR) has been identified as a new molecularly targeted entity, which is highly overexpressed on the surface of a spectrum of solid tumor cells, including ovarian, kidney, lung, brain, endometrial, colorectal, pancreatic, gastric, prostate, testicular, bladder, head and neck, breast, and non-small cell lung cancer. Folic acid conjugation is a novel approach for targeting FR-expressing tissues for personalized treatment. With the development of FRα-targeted therapies comes a concomitant prerequisite for reliable methods for the quantification of FRα tissue expression. Therefore, attaching a radioactive probe to folic acid to target diseased tissue has become a novel and powerful imaging technique. Currently available diagnostic tools frequently require invasive surgical biopsy. In contrast, the noninvasive single-photon emission computed tomography-based companion imaging agent, (99m)Tc-etarfolatide ((99m)Tc-EC20), is in development for use as a companion diagnostic with the FRα-targeted folate conjugate, vintafolide (EC145), to identify patients whose tumors express FRα. Vintafolide is a folic acid conjugate of Vinca alkaloid (desacetylvinblastine hydrazide) that targets FRα-expressing tumors, thereby disrupting microtubule polymerization. (99m)Tc-etarfolatide is taken up by FR-positive tumors and allows for noninvasive, whole-body monitoring of FRα expression status throughout treatment. The combination of vintafolide plus etarfolatide has been evaluated in three Phase 2 studies for the treatment of various solid tumors, including ovarian, endometrial, peritoneal, and platinum-resistant ovarian cancer, as well as lung cancer. Patients with FR-positive tumors, as identified by etarfolatide uptake, have had better clinical outcomes than patients with FR-negative tumors, indicating the potential of etarfolatide as a companion biomarker for predicting vintafolide response. Targeted therapies combined with a reliable companion diagnostic test represent a novel approach toward efficient personalized medicine for malignant and nonmalignant disorders. Furthermore, the recent availability of the crystal structures of FRα and FRβ in complex with folates and antifolates forms a realistic basis for the rational design and implementation of novel FR-targeted drugs for the treatment of cancer and inflammatory disorders.


International Journal of Cancer | 2007

Comparative preclinical activity of the folate-targeted Vinca alkaloid conjugates EC140 and EC145

Christopher P. Leamon; Joseph A. Reddy; Iontcho Radoslavov Vlahov; Elaine Westrick; Nikki Parker; Jeffrey S. Nicoson; Marilynn Vetzel

EC140 is a water soluble folate conjugate of desacetylvinblastine monohydrazide (DAVLBH), which is constructed with an endosome‐cleavable acyl hydrazone bond. This agent has proven to be active and specific against well established, subcutaneous folate receptor (FR)‐positive tumors in multiple animal models. Recent structure‐activity and optimization studies have yielded a disulfide bond‐containing counterpart to EC140, herein referred to as EC145. This new conjugate was found to retain high affinity for FR‐positive cells, and it produced specific, dose‐responsive activity in vitro. Comparative in vivo efficacy tests confirmed that, like EC140, EC145 displays activity against both syngeneic and xenograft tumor models. However, EC145 was found to be more active and better tolerated than EC140; hence, more durable complete responses were consistently observed in EC145‐treated tumor‐bearing animals. Furthermore, EC145 was not found to be active against a FR‐negative tumor model. Additional preclinical studies are therefore warranted to better understand EC145s breadth of activity against FR‐positive tumors.


Cancer Research | 2008

Folate Targeting Enables Durable and Specific Antitumor Responses from a Therapeutically Null Tubulysin B Analogue

Christopher P. Leamon; Joseph A. Reddy; Marilynn Vetzel; Ryan Dorton; Elaine Westrick; Nikki Parker; Yu Wang; Iontcho Radoslavov Vlahov

The membrane-bound high-affinity folate receptor (FR) is highly expressed on a wide range of primary and metastatic human cancers, such as those originating in ovary, lung, breast, endometrium, kidney, and brain. Because folate-linked conjugates bind to and become internalized within FR-expressing cells (similar to that of free folic acid), we explored the possibility of using the folate ligand to target a potent, semisynthetic analogue of the microtubule inhibitor tubulysin B to FR-enriched tumors. When tested in vitro, a novel folate conjugate, herein referred to as EC0305, was found to specifically inhibit the growth of a panel of FR-positive cell lines (IC50 range, 1-10 nmol/L) in a dose-dependent manner, whereas cells lacking FR expression were unaffected. The potency of EC0305 was also confirmed against a human KB xenograft-nu/nu mouse cancer model. Here, a brief three times per week, 2-week regimen yielded remarkable antitumor activity (100% tumor-free animals) without causing significant weight loss or major organ tissue degeneration. In contrast, antitumor activity was completely abolished in EC0305-treated animals that were co-dosed with an excess of a nontoxic folate-containing analogue, thereby confirming that the antitumor effect of this agent was mediated by FRs. The advantage provided by folate conjugation was further proved by the untargeted free drug, which was found to be completely inactive at both tolerable and highly toxic dose levels. Collectively, these results show that this potent antiproliferative tubulysin compound can be specifically delivered to FR-positive tumors to provide substantial therapeutic benefit using well-tolerable dosing regimens.


Cancer Chemotherapy and Pharmacology | 2006

Folate receptor specific anti-tumor activity of folate–mitomycin conjugates

Joseph A. Reddy; Elaine Westrick; Iontcho Radoslavov Vlahov; Stephen J. Howard; Hari Krishna R. Santhapuram; Christopher P. Leamon

Purpose: Folate receptor (FR) targeted drug conjugates were prepared by covalently attaching the vitamin folate, to the potent anticancer drug, mitomycin C (MMC). One such conjugate, called EC72, was synthesized with an intramolecular disulfide bond, and it was found to exhibit efficacious anti-tumor activity against FR-expressing M109 tumors in a manner that yielded no gross or microscopic toxicity, even to FR-positive kidneys. Methods: EC72’s specificity was demonstrated by two methods: (1) blocking EC72’s activity with an excess of co-administered folic acid (FA) in M109 tumor bearing mice and (2) the absence of therapeutic activity in mice bearing FR-negative tumors. The importance of having a cleavable bond in the conjugate was also exemplified, since EC110 (a folate–MMC conjugate constructed with a more resilient amide bond) failed to produce anti-M109 tumor activity. EC72’s therapeutic potential was found to decrease with respect to the increasing size of subcutaneous tumor. However, a combination therapy with paclitaxel reproducibly improved the anti-tumor efficacy relative to either agent alone at well tolerated dose levels and with no apparent increase in toxicity. A more advanced folate–MMC conjugate was also synthesized in an effort to improve activity. Thus, EC118, a molecule constructed with both a reducible disulfide bond and an acid-labile hydrazone bond in the linker region, was tested and found to produce a significantly greater number of tumor regressions of more established M109 tumors than that achieved with EC72. Conclusion: Overall, these data indicate that folate-targeted drug therapy alone, or in combination with paclitaxel, may be a novel and effective clinical approach towards treating FR-positive cancers.


Cancer Research | 2007

Folate Receptor–Specific Antitumor Activity of EC131, a Folate-Maytansinoid Conjugate

Joseph A. Reddy; Elaine Westrick; Hari Krishna R. Santhapuram; Stephen J. Howard; Michael L. Miller; Marilynn Vetzel; Iontcho Radoslavov Vlahov; Ravi V. J. Chari; Victor S. Goldmacher; Christopher P. Leamon

EC131, a new folate receptor (FR)-targeted drug conjugate, was prepared by covalently attaching the vitamin folic acid (FA) to a potent microtubule-inhibiting agent, maytansinoid DM1, via an intramolecular disulfide bond. When tested on cells in culture, EC131 was found to retain high affinity for FR-positive cells and to provide FR-specific cytotoxicity with an IC(50) in the low nanomolar range. The activity of EC131 was completely blocked in the presence of an excess of free FA, and no activity was detected against FR-negative cells. When evaluated against s.c. FR-positive M109 tumors in BALB/c mice, EC131 showed marked antitumor efficacy. Furthermore, this therapeutic effect occurred in the apparent absence of weight loss or noticeable organ tissue degeneration. In contrast, no significant antitumor activity was observed in EC131-treated animals that were codosed with an excess of FA, thus demonstrating the targeted specificity of the in vivo activity. EC131 also showed marked antitumor activity against FR-positive human KB tumors, but not against FR-negative A549 tumors, in nude mice with no evidence of systemic toxicity during or after the therapy. In contrast, therapy with the free maytansinoid drug (in the form of DM1-S-Me) proved not to be effective against the KB model when administered at its maximum tolerated dose (MTD). Taken together, these results indicate that EC131 is a highly potent agent capable of producing therapeutic benefit in murine tumor models at sub-MTD levels.

Collaboration


Dive into the Christopher P. Leamon's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge