Network


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

Hotspot


Dive into the research topics where Lisa Crocker is active.

Publication


Featured researches published by Lisa Crocker.


Cancer Research | 2008

Targeting HER2-positive breast cancer with trastuzumab-DM1, an antibody-cytotoxic drug conjugate.

Gail Lewis Phillips; Guangmin Li; Debra L. Dugger; Lisa Crocker; Kathryn Parsons; Elaine Mai; Walter A. Blattler; John M. Lambert; Ravi V. J. Chari; Robert J. Lutz; Wai Lee T. Wong; Frederic S. Jacobson; Hartmut Koeppen; Ralph Schwall; Sara R. Kenkare-Mitra; Susan D. Spencer; Mark X. Sliwkowski

HER2 is a validated target in breast cancer therapy. Two drugs are currently approved for HER2-positive breast cancer: trastuzumab (Herceptin), introduced in 1998, and lapatinib (Tykerb), in 2007. Despite these advances, some patients progress through therapy and succumb to their disease. A variation on antibody-targeted therapy is utilization of antibodies to deliver cytotoxic agents specifically to antigen-expressing tumors. We determined in vitro and in vivo efficacy, pharmacokinetics, and toxicity of trastuzumab-maytansinoid (microtubule-depolymerizing agents) conjugates using disulfide and thioether linkers. Antiproliferative effects of trastuzumab-maytansinoid conjugates were evaluated on cultured normal and tumor cells. In vivo activity was determined in mouse breast cancer models, and toxicity was assessed in rats as measured by body weight loss. Surprisingly, trastuzumab linked to DM1 through a nonreducible thioether linkage (SMCC), displayed superior activity compared with unconjugated trastuzumab or trastuzumab linked to other maytansinoids through disulfide linkers. Serum concentrations of trastuzumab-MCC-DM1 remained elevated compared with other conjugates, and toxicity in rats was negligible compared with free DM1 or trastuzumab linked to DM1 through a reducible linker. Potent activity was observed on all HER2-overexpressing tumor cells, whereas nontransformed cells and tumor cell lines with normal HER2 expression were unaffected. In addition, trastuzumab-DM1 was active on HER2-overexpressing, trastuzumab-refractory tumors. In summary, trastuzumab-DM1 shows greater activity compared with nonconjugated trastuzumab while maintaining selectivity for HER2-overexpressing tumor cells. Because trastuzumab linked to DM1 through a nonreducible linker offers improved efficacy and pharmacokinetics and reduced toxicity over the reducible disulfide linkers evaluated, trastuzumab-MCC-DM1 was selected for clinical development.


Cancer Research | 2008

A Central Role for HER3 in HER2-Amplified Breast Cancer: Implications for Targeted Therapy

Si Tuen Lee-Hoeflich; Lisa Crocker; Evelyn Yao; Thinh Pham; Xander Munroe; Klaus P. Hoeflich; Mark X. Sliwkowski; Howard M. Stern

Epidermal growth factor receptor (EGFR) and HER3 each form heterodimers with HER2 and have independently been implicated as key coreceptors that drive HER2-amplified breast cancer. Some studies suggest a dominant role for EGFR, a notion of renewed interest given the development of dual HER2/EGFR small-molecule inhibitors. Other studies point to HER3 as the primary coreceptor. To clarify the relative contributions of EGFR and HER3 to HER2 signaling, we studied receptor knockdown via small interfering RNA technology across a panel of six HER2-overexpressing cell lines. Interestingly, HER3 was as critical as HER2 for maintaining cell proliferation in most cell lines, whereas EGFR was dispensable. Induction of HER3 knockdown in the HER2-overexpressing BT474M1 cell line was found to inhibit growth in three-dimensional culture and induce rapid tumor regression of in vivo xenografts. Furthermore, preferential phosphorylation of HER3, but not EGFR, was observed in HER2-amplified breast cancer tissues. Given these data suggesting HER3 as an important therapeutic target, we examined the activity of pertuzumab, a HER2 antibody that inhibits HER3 signaling by blocking ligand-induced HER2/HER3 heterodimerization. Pertuzumab inhibited ligand-dependent morphogenesis in three-dimensional culture and induced tumor regression in the heregulin-dependent MDA-MB-175 xenograft model. Importantly, these activities of pertuzumab were distinct from those of trastuzumab, a monoclonal antibody currently used for treatment of HER2-amplified breast cancer patients. Our data suggest that inhibition of HER3 may be more clinically relevant than inhibition of EGFR in HER2-amplified breast cancer and also suggest that adding pertuzumab to trastuzumab may augment therapeutic benefit by blocking HER2/HER3 signaling.


Cancer Research | 2010

Superior In vivo Efficacy of Afucosylated Trastuzumab in the Treatment of HER2-Amplified Breast Cancer

Teemu T. Junttila; Kathryn Parsons; Christine Olsson; Yanmei Lu; Yan Xin; Julie Theriault; Lisa Crocker; Oliver Pabonan; Tomasz Baginski; Gloria Meng; Klara Totpal; Robert F. Kelley; Mark X. Sliwkowski

The enhancement of immune effector functions has been proposed as a potential strategy for increasing the efficacy of therapeutic antibodies. Here, we show that removing fucose from trastuzumab (Herceptin) increased its binding to FcgammaRIIIa, enhanced antibody-dependent cell-mediated cytotoxicity, and more than doubled the median progression-free survival when compared with conventional trastuzumab in treating preclinical models of HER2-amplified breast cancer. Our results show that afucosylated trastuzumab has superior efficacy in treating in vivo models of HER2-amplified breast cancer and support the development of effector function-enhanced antibodies for solid tumor therapy.


Clinical Cancer Research | 2014

Dual Targeting of HER2-Positive Cancer with Trastuzumab Emtansine and Pertuzumab: Critical Role for Neuregulin Blockade in Antitumor Response to Combination Therapy

Gail Lewis Phillips; Carter Fields; Guangmin Li; Donald Dowbenko; Gabriele Schaefer; Kathy D. Miller; Fabrice Andre; Howard A. Burris; Kathy S. Albain; Nadia Harbeck; V. Dieras; Diana Crivellari; Liang Fang; Ellie Guardino; Steven Olsen; Lisa Crocker; Mark X. Sliwkowski

Purpose: Targeting HER2 with multiple HER2-directed therapies represents a promising area of treatment for HER2-positive cancers. We investigated combining the HER2-directed antibody–drug conjugate trastuzumab emtansine (T-DM1) with the HER2 dimerization inhibitor pertuzumab (Perjeta). Experimental Design: Drug combination studies with T-DM1 and pertuzumab were performed on cultured tumor cells and in mouse xenograft models of HER2-amplified cancer. In patients with HER2-positive locally advanced or metastatic breast cancer (mBC), T-DM1 was dose-escalated with a fixed standard pertuzumab dose in a 3+3 phase Ib/II study design. Results: Treatment of HER2-overexpressing tumor cells in vitro with T-DM1 plus pertuzumab resulted in synergistic inhibition of cell proliferation and induction of apoptotic cell death. The presence of the HER3 ligand, heregulin (NRG-1β), reduced the cytotoxic activity of T-DM1 in a subset of breast cancer lines; this effect was reversed by the addition of pertuzumab. Results from mouse xenograft models showed enhanced antitumor efficacy with T-DM1 and pertuzumab resulting from the unique antitumor activities of each agent. In patients with mBC previously treated with trastuzumab, lapatinib, and chemotherapy, T-DM1 could be dosed at the maximum tolerated dose (MTD; 3.6 mg/kg every 3 weeks) with standard dose pertuzumab. Adverse events were mostly grade 1 and 2, with indications of clinical activity. Conclusions: Dual targeting of HER2 with the combination of T-DM1 and pertuzumab in cell culture and mouse xenograft models resulted in enhanced antitumor activity. In patients, this combination showed an encouraging safety and tolerability profile with preliminary evidence of efficacy. Clin Cancer Res; 20(2); 456–68. ©2013 AACR.


Journal of Pharmacokinetics and Pharmacodynamics | 2010

Modeling the efficacy of trastuzumab-DM1, an antibody drug conjugate, in mice

Nelson L. Jumbe; Yan Xin; Douglas D. Leipold; Lisa Crocker; Debra L. Dugger; Elaine Mai; Mark X. Sliwkowski; Paul J. Fielder; Jay Tibbitts

Trastuzumab-DM1 (T-DM1) is a novel antibody–drug conjugate under investigation for the treatment of human epidermal growth factor receptor 2 (HER2)-positive metastatic breast cancer. One challenge in oncologic drug development is determining the optimal dose and treatment schedule. A novel dose regimen-finding strategy was developed for T-DM1 using experimental data and pharmacokinetic/pharmacodynamic modeling. To characterize the disposition of T-DM1, pharmacokinetic studies were conducted in athymic nude and beige nude mice. The pharmacokinetics of T-DM1 were described well by a two-compartment model. Tumor response data were obtained from single-dose, multiple-dose and time–dose-fractionation studies of T-DM1 in animal models of HER2-positive breast cancer, specifically engineered to be insensitive to trastuzumab. A sequential population-based pharmacokinetic/pharmacodynamic modeling approach was developed to describe the anti-tumor activity of T-DM1. A cell-cycle-phase nonspecific tumor cell kill model incorporating transit compartments captured well the features of tumor growth and the activity of T-DM1. Key findings of the model were that tumor cell growth rate played a significant role in the sensitivity of tumors to T-DM1; anti-tumor activity was schedule independent; and tumor response was linked to the ratio of exposure to a concentration required for tumor stasis.


Science Translational Medicine | 2013

Blocking NRG1 and Other Ligand-Mediated Her4 Signaling Enhances the Magnitude and Duration of the Chemotherapeutic Response of Non–Small Cell Lung Cancer

Ganapati V. Hegde; Cecile de la Cruz; Cecilia Chiu; Navneet Alag; Gabriele Schaefer; Lisa Crocker; Sarajane Ross; David M. Goldenberg; Mark Merchant; Janet Tien; Lily Shao; Leslie Roth; Siao-Ping Tsai; Scott Stawicki; Zhaoyu Jin; Shelby K. Wyatt; Richard A. D. Carano; Yanyan Zheng; E. Alejandro Sweet-Cordero; Yan Wu; Erica Jackson

Inhibition of Her4 signaling enhances the response to chemotherapy and delays tumor regrowth after cessation of treatment. Regaining the Yellow Jersey Professional sports—from cycling to football and even baseball—are now cracking down on doping. The use of performance-enhancing drugs is thought to give an unfair advantage, and regulatory agencies are trying to return everyone to even ground. But whereas athletes who dope become pariahs, in some fights it’s better not to play fair. Now, Hegde et al. suggest a way to enhance chemotherapy in the fight against non–small cell lung cancer (NSCLC). Chemotherapy is a first-line treatment for NSCLC but, in some cases, cannot either adequately remove the tumor or prevent recurrence. The authors use multiple models of NSCLC and find that residual tumor cells after chemotherapy express high levels of neuregulin 1 (NRG1), which is a ligand for human epidermal growth factor receptor 3 and 4 (HER3/4). Inhibited NRG1 signaling had only variable effects on primary tumor growth, but significantly enhanced the magnitude and duration of tumor response to chemotherapy. NRG1 inhibition in combination with chemotherapy greatly impeded relapse. Although this combination remains to be tested in the clinic, this study suggests that when it comes to a competition between NSCLC and chemotherapy, all’s fair. Although standard chemotherapies are commonly used to treat most types of solid tumors, such treatment often results in inadequate response to, or relapse after, therapy. This is particularly relevant for lung cancer because most patients are diagnosed with advanced-stage disease and are treated with frontline chemotherapy. By studying the residual tumor cells that remain after chemotherapy in several in vivo non–small cell lung cancer models, we found that these cells have increased levels of human epidermal growth factor receptor (HER) signaling due, in part, to the enrichment of a preexisting NRG1HI subpopulation. Neuregulin 1 (NRG1) signaling in these models can be mediated by either the HER3 or HER4 receptor, resulting in the differential activation of downstream effectors. Inhibition of NRG1 signaling inhibits primary tumor growth and enhances the magnitude and duration of the response to chemotherapy. Moreover, we show that inhibition of ligand-mediated Her4 signaling impedes disease relapse in cases where NRG1 inhibition is insufficient. These findings demonstrate that ligand-dependent Her4 signaling plays an important role in disease relapse.


Journal of Medicinal Chemistry | 2013

Fragment-Based Identification of Amides Derived from trans-2-(Pyridin-3-yl)cyclopropanecarboxylic Acid as Potent Inhibitors of Human Nicotinamide Phosphoribosyltransferase (NAMPT)

Anthony M. Giannetti; Xiaozhang Zheng; Nicholas J. Skelton; Weiru Wang; Brandon J. Bravo; Kenneth W. Bair; Timm Baumeister; Eric Cheng; Lisa Crocker; Yezhen Feng; Janet Gunzner-Toste; Yen-Ching Ho; Rongbao Hua; Bianca M. Liederer; Yongbo Liu; Xiaolei Ma; Thomas O’Brien; Jason Oeh; Deepak Sampath; Youming Shen; Chengcheng Wang; Leslie Wang; Hongxing Wu; Yang Xiao; Po-wai Yuen; Mark Zak; Guiling Zhao; Qiang Zhao; Peter S. Dragovich

Potent, trans-2-(pyridin-3-yl)cyclopropanecarboxamide-containing inhibitors of the human nicotinamide phosphoribosyltransferase (NAMPT) enzyme were identified using fragment-based screening and structure-based design techniques. Multiple crystal structures were obtained of initial fragment leads, and this structural information was utilized to improve the biochemical and cell-based potency of the associated molecules. Many of the optimized compounds exhibited nanomolar antiproliferative activities against human tumor lines in in vitro cell culture experiments. In a key example, a fragment lead (13, KD = 51 μM) was elaborated into a potent NAMPT inhibitor (39, NAMPT IC50 = 0.0051 μM, A2780 cell culture IC50 = 0.000 49 μM) which demonstrated encouraging in vivo efficacy in an HT-1080 mouse xenograft tumor model.


Molecular Pharmaceutics | 2015

An Anti-B7-H4 Antibody–Drug Conjugate for the Treatment of Breast Cancer

Steven R. Leong; Wei Ching Liang; Yan Wu; Lisa Crocker; Eric Cheng; Deepak Sampath; Rachana Ohri; Helga Raab; Philip E. Hass; Thinh Pham; Ron Firestein; Dongwei Li; Melissa Schutten; Nicola J. Stagg; Annie Ogasawara; Neelima Koppada; Leslie Roth; Simon Williams; Byoung Chul Lee; Cecile Chalouni; Ivan Peng; Jason DeVoss; Jarrod Tremayne; Paul Polakis; Andrew G. Polson

B7-H4 has been implicated in cancers of the female reproductive system and investigated for its possible use as a biomarker for cancer, but there are no preclinical studies to demonstrate that B7-H4 is a molecular target for therapeutic intervention of cancer. We provide evidence that the prevalence and expression levels of B7-H4 are high in different subtypes of breast cancer and that only a few normal tissues express B7-H4 on the cell membrane. These profiles of low normal expression and upregulation in cancer provide an opportunity for the use of antibody-drug conjugates (ADCs), cytotoxic drugs chemically linked to antibodies, for the treatment of B7-H4 positive cancers. We have developed an ADC specific to B7-H4 that uses a linker drug consisting of a potent antimitotic, monomethyl auristatin E (MMAE), linked to engineered cysteines (THIOMAB) via a protease labile linker. We will refer to ADCs that use the THIOMAB format as TDCs to help distinguish the format from standard MC-vc-MMAE ADCs that are conjugated to the interchain disulfide bonds. Anti-B7-H4 (h1D11)-MC-vc-PAB-MMAE (h1D11 TDC) produced durable tumor regression in cell line and patient-derived xenograft models of triple-negative breast cancer. It also binds rat B7-H4 with similar affinity to human and allowed us to test for target dependent toxicity in rats. We found that our anti-B7-H4 TDC has toxicity findings similar to untargeted TDC. Our results validate B7-H4 as an ADC target for breast cancer and support the possible use of this TDC in the treatment of B7-H4(+) breast cancer.


Clinical Cancer Research | 2017

Single-Cell Dynamics Determines Response to CDK4/6 Inhibition in Triple-Negative Breast Cancer.

Uzma Asghar; Alexis R. Barr; Ros Cutts; Matthew Beaney; Irina S. Babina; Deepak Sampath; Jennifer Giltnane; Jennifer A. Lacap; Lisa Crocker; Amy Young; Alex Pearson; Maria Teresa Herrera-Abreu; Chris Bakal; Nicholas C. Turner

Purpose: Triple-negative breast cancer (TNBC) is a heterogeneous subgroup of breast cancer that is associated with a poor prognosis. We evaluated the activity of CDK4/6 inhibitors across the TNBC subtypes and investigated mechanisms of sensitivity. Experimental Design: A panel of cell lines representative of TNBC was tested for in vitro and in vivo sensitivity to CDK4/6 inhibition. A fluorescent CDK2 activity reporter was used for single-cell analysis in conjunction with time-lapse imaging. Results: The luminal androgen receptor (LAR) subtype of TNBC was highly sensitive to CDK4/6 inhibition both in vitro (P < 0.001 LAR vs. basal-like) and in vivo in MDA-MB-453 LAR cell line xenografts. Single-cell analysis of CDK2 activity demonstrated differences in cell-cycle dynamics between LAR and basal-like cells. Palbociclib-sensitive LAR cells exit mitosis with low levels of CDK2 activity, into a quiescent state that requires CDK4/6 activity for cell-cycle reentry. Palbociclib-resistant basal-like cells exit mitosis directly into a proliferative state, with high levels of CDK2 activity, bypassing the restriction point and the requirement for CDK4/6 activity. High CDK2 activity after mitosis is driven by temporal deregulation of cyclin E1 expression. CDK4/6 inhibitors were synergistic with PI3 kinase inhibitors in PIK3CA-mutant TNBC cell lines, extending CDK4/6 inhibitor sensitivity to additional TNBC subtypes. Conclusions: Cell-cycle dynamics determine the response to CDK4/6 inhibition in TNBC. CDK4/6 inhibitors, alone and in combination, are a novel therapeutic strategy for specific subgroups of TNBC. Clin Cancer Res; 23(18); 5561–72. ©2017 AACR.


PLOS ONE | 2014

Structural Basis for Resistance to Diverse Classes of NAMPT Inhibitors.

Weiru Wang; Kristi Elkins; Angela Oh; Yen-Ching Ho; Jiansheng Wu; Hong Li; Yang Xiao; Mandy Kwong; Mary Coons; Bobby Brillantes; Eric Cheng; Lisa Crocker; Peter S. Dragovich; Deepak Sampath; Xiaozhang Zheng; Kenneth W. Bair; Thomas O'Brien; Lisa D. Belmont

Inhibiting NAD biosynthesis by blocking the function of nicotinamide phosphoribosyl transferase (NAMPT) is an attractive therapeutic strategy for targeting tumor metabolism. However, the development of drug resistance commonly limits the efficacy of cancer therapeutics. This study identifies mutations in NAMPT that confer resistance to a novel NAMPT inhibitor, GNE-618, in cell culture and in vivo, thus demonstrating that the cytotoxicity of GNE-618 is on target. We determine the crystal structures of six NAMPT mutants in the apo form and in complex with various inhibitors and use cellular, biochemical and structural data to elucidate two resistance mechanisms. One is the surprising finding of allosteric modulation by mutation of residue Ser165, resulting in unwinding of an α-helix that binds the NAMPT substrate 5-phosphoribosyl-1-pyrophosphate (PRPP). The other mechanism is orthosteric blocking of inhibitor binding by mutations of Gly217. Furthermore, by evaluating a panel of diverse small molecule inhibitors, we unravel inhibitor structure activity relationships on the mutant enzymes. These results provide valuable insights into the design of next generation NAMPT inhibitors that offer improved therapeutic potential by evading certain mechanisms of resistance.

Collaboration


Dive into the Lisa Crocker's collaboration.

Researchain Logo
Decentralizing Knowledge