Network


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

Hotspot


Dive into the research topics where Rachana Ohri is active.

Publication


Featured researches published by Rachana Ohri.


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.


Journal of Biological Chemistry | 2012

The melanosomal protein PMEL17 as a target for antibody drug conjugate therapy in melanoma

Youjun Chen; Cecile Chalouni; Christine Tan; Robyn Clark; Rayna Venook; Rachana Ohri; Helga Raab; Ron Firestein; William Mallet; Paul Polakis

Background: A search for cell surface proteins amenable to antibody drug conjugate (ADC) therapy was performed. Results: Expression of PMEL17 was highly restricted to melanoma cells, and an ADC directed against it was efficacious. Conclusion: PMEL17 is an attractive target for ADC therapy in melanoma. Significance: Intracellular transmembrane proteins that transit the cell surface represent a new class of targets for ADCs. Melanocytes uniquely express specialized genes required for pigment formation, some of which are maintained following their transformation to melanoma. Here we exploit this property to selectively target melanoma with an antibody drug conjugate (ADC) specific to PMEL17, the product of the SILV pigment-forming gene. We describe new PMEL17 antibodies that detect the endogenous protein. These antibodies help define the secretory fate of PMEL17 and demonstrate its utility as an ADC target. Although newly synthesized PMEL17 is ultimately routed to the melanosome, we find substantial amounts accessible to our antibodies at the cell surface that undergo internalization and routing to a LAMP1-enriched, lysosome-related organelle. Accordingly, an ADC reactive with PMEL17 exhibits target-dependent tumor cell killing in vitro and in vivo.


Molecular Cancer Therapeutics | 2017

Modulating Therapeutic Activity and Toxicity of Pyrrolobenzodiazepine Antibody-Drug Conjugates with Self-Immolative Disulfide Linkers

Thomas H. Pillow; Melissa Schutten; Shang-Fan Yu; Rachana Ohri; Jack Sadowsky; Kirsten Achilles Poon; Willy Solis; Fiona Zhong; Geoffrey Del Rosario; Mary Ann T. Go; Jeffrey Lau; Sharon Yee; Jintang He; Luna Liu; Carl Ng; Keyang Xu; Douglas D. Leipold; Amrita V. Kamath; Donglu Zhang; Luke Masterson; Stephen J. Gregson; Philip W. Howard; Fan Fang; Jinhua Chen; Janet Gunzner-Toste; Katherine K. Kozak; Susan D. Spencer; Paul Polakis; Andrew G. Polson; John A. Flygare

A novel disulfide linker was designed to enable a direct connection between cytotoxic pyrrolobenzodiazepine (PBD) drugs and the cysteine on a targeting antibody for use in antibody–drug conjugates (ADCs). ADCs composed of a cysteine-engineered antibody were armed with a PBD using a self-immolative disulfide linker. Both the chemical linker and the antibody site were optimized for this new bioconjugation strategy to provide a highly stable and efficacious ADC. This novel disulfide ADC was compared with a conjugate containing the same PBD drug, but attached to the antibody via a peptide linker. Both ADCs had similar efficacy in mice bearing human tumor xenografts. Safety studies in rats revealed that the disulfide-linked ADC had a higher MTD than the peptide-linked ADC. Overall, these data suggest that the novel self-immolative disulfide linker represents a valuable way to construct ADCs with equivalent efficacy and improved safety. Mol Cancer Ther; 16(5); 871–8. ©2017 AACR.


Bioconjugate Chemistry | 2018

High-Throughput Cysteine Scanning To Identify Stable Antibody Conjugation Sites for Maleimide- and Disulfide-Based Linkers

Rachana Ohri; Sunil Bhakta; Aimee Fourie-O’Donohue; Josefa dela Cruz-Chuh; Siao Ping Tsai; Ryan Cook; Binqing Wei; Carl Ng; Athena W. Wong; Aaron B. Bos; Farzam Farahi; Jiten Bhakta; Thomas H. Pillow; Helga Raab; Richard Vandlen; Paul Polakis; Yichin Liu; Hans Erickson; Jagath R. Junutula; Katherine R. Kozak

THIOMAB antibody technology utilizes cysteine residues engineered onto an antibody to allow for site-specific conjugation. The technology has enabled the exploration of different attachment sites on the antibody in combination with small molecules, peptides, or proteins to yield antibody conjugates with unique properties. As reported previously ( Shen , B. Q. , et al. ( 2012 ) Nat. Biotechnol. 30 , 184 - 189 ; Pillow , T. H. , et al. ( 2017 ) Chem. Sci. 8 , 366 - 370 ), the specific location of the site of conjugation on an antibody can impact the stability of the linkage to the engineered cysteine for both thio-succinimide and disulfide bonds. High stability of the linkage is usually desired to maximize the delivery of the cargo to the intended target. In the current study, cysteines were individually substituted into every position of the anti-HER2 antibody (trastuzumab), and the stabilities of drug conjugations at those sites were evaluated. We screened a total of 648 THIOMAB antibody-drug conjugates, each generated from a trastuzamab prepared by sequentially mutating non-cysteine amino acids in the light and heavy chains to cysteine. Each THIOMAB antibody variant was conjugated to either maleimidocaproyl-valine-citrulline-p-aminobenzyloxycarbonyl-monomethyl auristatin E (MC-vc-PAB-MMAE) or pyridyl disulfide monomethyl auristatin E (PDS-MMAE) using a high-throughput, on-bead conjugation and purification method. Greater than 50% of the THIOMAB antibody variants were successfully conjugated to both MMAE derivatives with a drug to antibody ratio (DAR) of >0.5 and <50% aggregation. The relative in vitro plasma stabilities for approximately 750 conjugates were assessed using enzyme-linked immunosorbent assays, and stable sites were confirmed with affinity-capture LC/MS-based detection methods. Highly stable conjugation sites for the two types of MMAE derivatives were identified on both the heavy and light chains. Although the stabilities of maleimide conjugates were shown to be greater than those of the disulfide conjugates, many sites were identified that were stable for both. Furthermore, in vitro stabilities of selected stable sites translated across different cytotoxic payloads and different target antibodies as well as to in vivo stability.


Analytical Chemistry | 2017

High-Resolution Accurate-Mass Mass Spectrometry Enabling In-Depth Characterization of in Vivo Biotransformations for Intact Antibody-Drug Conjugates

Jintang He; Dian Su; Carl Ng; Luna Liu; Shang-Fan Yu; Thomas H. Pillow; Geoffrey Del Rosario; Martine Darwish; Byoung-Chul Lee; Rachana Ohri; Hongxiang Zhou; Xueji Wang; Jiawei Lu; Surinder Kaur; Keyang Xu

Antibody-drug conjugates (ADCs) represent a promising class of therapeutics for the targeted delivery of highly potent cytotoxic drugs to tumor cells to improve bioactivity while minimizing side effects. ADCs are composed of both small and large molecules and therefore have complex molecular structures. In vivo biotransformations may further increase the complexity of ADCs, representing a unique challenge for bioanalytical assays. Quadrupole-time-of-flight mass spectrometry (Q-TOF MS) with electrospray ionization has been widely used for characterization of intact ADCs. However, interpretation of ADC biotransformations with small mass changes, for the intact molecule, remains a limitation due to the insufficient mass resolution and accuracy of Q-TOF MS. Here, we have investigated in vivo biotransformations of multiple site-specific THIOMAB antibody-drug conjugates (TDCs), in the intact form, using a high-resolution, accurate-mass (HR/AM) MS approach. Compared with conventional Q-TOF MS, HR/AM Orbitrap MS enabled more comprehensive identification of ADC biotransformations. It was particularly beneficial for characterizing ADC modifications with small mass changes such as partial drug loss and hydrolysis. This strategy has significantly enhanced our capability to elucidate ADC biotransformations and help understand ADC efficacy and safety in vivo.


Journal of Medicinal Chemistry | 2017

Pyrrolobenzodiazepine Dimer Antibody-Drug Conjugates: Synthesis and Evaluation of Non-Cleavable Drug-Linkers.

Stephen J. Gregson; Luke Masterson; Binqing Wei; Thomas H. Pillow; Susan D. Spencer; Gyoung-Dong Kang; Shang-Fan Yu; Helga Raab; Jeffrey Lau; Guangmin Li; Gail Lewis Phillips; Janet Gunzner-Toste; Brian Safina; Rachana Ohri; Martine Darwish; Katherine R. Kozak; Josefa dela Cruz-Chuh; Andrew Polson; John A. Flygare; Philip W. Howard

Three rationally designed pyrrolobenzodiazepine (PBD) drug-linkers have been synthesized via intermediate 19 for use in antibody-drug conjugates (ADCs). They lack a cleavable trigger in the linker and consist of a maleimide for cysteine antibody conjugation, a hydrophilic spacer, and either an alkyne (6), triazole (7), or piperazine (8) link to the PBD. In vitro IC50 values were 11-48 ng/mL in HER2 3+ SK-BR-3 and KPL-4 (7 inactive) for the anti-HER2 ADCs (HER2 0 MCF7, all inactive) and 0.10-1.73 μg/mL (7 inactive) in CD22 3+ BJAB and WSU-DLCL2 for anti-CD22 ADCs (CD22 0 Jurkat, all inactive at low doses). In vivo antitumor efficacy for the anti-HER2 ADCs in Founder 5 was observed with tumor stasis at 0.5-1 mg/kg, 1 mg/kg, and 3-6 mg/kg for 6, 8, and 7, respectively. Tumor stasis at 2 mg/kg was observed for anti-CD22 6 in WSU-DLCL2. In summary, noncleavable PBD-ADCs exhibit potent activity, particularly in HER2 models.


Bioconjugate Chemistry | 2017

Attachment Site Cysteine Thiol pKa Is a Key Driver for Site-Dependent Stability of THIOMAB Antibody–Drug Conjugates

Breanna S. Vollmar; Binqing Wei; Rachana Ohri; Jianhui Zhou; Jintang He; Shang-Fan Yu; Douglas D. Leipold; Ely Cosino; Sharon Yee; Aimee Fourie-O’Donohue; Guangmin Li; Gail Lewis Phillips; Katherine R. Kozak; Amrita Kamath; Keyang Xu; Genee Lee; Greg A. Lazar; Hans K. Erickson

The incorporation of cysteines into antibodies by mutagenesis allows for the direct conjugation of small molecules to specific sites on the antibody via disulfide bonds. The stability of the disulfide bond linkage between the small molecule and the antibody is highly dependent on the location of the engineered cysteine in either the heavy chain (HC) or the light chain (LC) of the antibody. Here, we explore the basis for this site-dependent stability. We evaluated the in vivo efficacy and pharmacokinetics of five different cysteine mutants of trastuzumab conjugated to a pyrrolobenzodiazepine (PBD) via disulfide bonds. A significant correlation was observed between disulfide stability and efficacy for the conjugates. We hypothesized that the observed site-dependent stability of the disulfide-linked conjugates could be due to differences in the attachment site cysteine thiol pKa. We measured the cysteine thiol pKa using isothermal titration calorimetry (ITC) and found that the variants with the highest thiol pKa (LC K149C and HC A140C) were found to yield the conjugates with the greatest in vivo stability. Guided by homology modeling, we identified several mutations adjacent to LC K149C that reduced the cysteine thiol pKa and, thus, decreased the in vivo stability of the disulfide-linked PBD conjugated to LC K149C. We also present results suggesting that the high thiol pKa of LC K149C is responsible for the sustained circulation stability of LC K149C TDCs utilizing a maleimide-based linker. Taken together, our results provide evidence that the site-dependent stability of cys-engineered antibody-drug conjugates may be explained by interactions between the engineered cysteine and the local protein environment that serves to modulate the side-chain thiol pKa. The influence of cysteine thiol pKa on stability and efficacy offers a new parameter for the optimization of ADCs that utilize cysteine engineering.


ACS Medicinal Chemistry Letters | 2017

Stabilizing a Tubulysin Antibody–Drug Conjugate To Enable Activity Against Multidrug-Resistant Tumors

Leanna Staben; Shang-Fan Yu; Jinhua Chen; Gang Yan; Zijin Xu; Geoffrey Del Rosario; Jeffrey Lau; Luna Liu; Jun Guo; Bing Zheng; Josefa dela Cruz-Chuh; Byoung-Chul Lee; Rachana Ohri; Wenwen Cai; Hongxiang Zhou; Katherine R. Kozak; Keyang Xu; Gail Lewis Phillips; Jiawei Lu; John S. Wai; Andrew Polson; Thomas H. Pillow

The tubulysins are promising anticancer cytotoxic agents due to the clinical validation of their mechanism of action (microtubule inhibition) and their particular activity against multidrug-resistant tumor cells. Yet their high potency and subsequent systemic toxicity make them prime candidates for targeted therapy, particularly in the form of antibody-drug conjugates (ADCs). Here we report a strategy to prepare stable and bioreversible conjugates of tubulysins to antibodies without loss of activity. A peptide trigger along with a quaternary ammonium salt linker connection to the tertiary amine of tubulysin provided ADCs that were potent in vitro. However, we observed metabolism of a critical acetate ester of the drug in vivo, resulting in diminished conjugate activity. We were able to circumvent this metabolic liability with the judicious choice of a propyl ether replacement. This modified tubulysin ADC was stable and effective against multidrug-resistant lymphoma cell lines and tumors.


Bioconjugate Chemistry | 2018

Modulating Antibody–Drug Conjugate Payload Metabolism by Conjugation Site and Linker Modification

Dian Su; Katherine R. Kozak; Jack Sadowsky; Shang-Fan Yu; Aimee Fourie-O’Donohue; Christopher Nelson; Richard Vandlen; Rachana Ohri; Luna Liu; Carl Ng; Jintang He; Helen Davis; Jeff Lau; Geoffrey Del Rosario; Ely Cosino; Josefa dela Cruz-Chuh; Yong Ma; Donglu Zhang; Martine Darwish; Wenwen Cai; Chunjiao Chen; Hongxiang Zhou; Jiawei Lu; Yichin Liu; Surinder Kaur; Keyang Xu; Thomas H. Pillow

Previous investigations on antibody-drug conjugate (ADC) stability have focused on drug release by linker-deconjugation due to the relatively stable payloads such as maytansines. Recent development of ADCs has been focused on exploring technologies to produce homogeneous ADCs and new classes of payloads to expand the mechanisms of action of the delivered drugs. Certain new ADC payloads could undergo metabolism in circulation while attached to antibodies and thus affect ADC stability, pharmacokinetics, and efficacy and toxicity profiles. Herein, we investigate payload stability specifically and seek general guidelines to address payload metabolism and therefore increase the overall ADC stability. Investigation was performed on various payloads with different functionalities (e.g., PNU-159682 analog, tubulysin, cryptophycin, and taxoid) using different conjugation sites (HC-A118C, LC-K149C, and HC-A140C) on THIOMAB antibodies. We were able to reduce metabolism and inactivation of a broad range of payloads of THIOMAB antibody-drug conjugates by employing optimal conjugation sites (LC-K149C and HC-A140C). Additionally, further payload stability was achieved by optimizing the linkers. Coupling relatively stable sites with optimized linkers provided optimal stability and reduction of payloads metabolism in circulation in vivo.


Molecular Cancer Therapeutics | 2017

An anti-GDNF Family Receptor Alpha 1(GFRA1) Antibody-Drug Conjugate for the Treatment of Hormone Receptor-Positive Breast Cancer

Sunil Bhakta; Lisa Crocker; Yvonne Chen; Meredith Hazen; Melissa Schutten; Dongwei Li; Coenraad Kuijl; Rachana Ohri; Fiona Zhong; Kirsten Achilles Poon; Mary Ann T. Go; Eric Cheng; Robert Piskol; Ron Firestein; Aimee Fourie-O'Donohue; Katherine R. Kozak; Helga Raab; Jo-Anne Hongo; Deepak Sampath; Mark S. Dennis; Richard H. Scheller; Paul Polakis; Jagath R. Junutula

Luminal A (hormone receptor-positive) breast cancer constitutes 70% of total breast cancer patients. In an attempt to develop a targeted therapeutic for this cancer indication, we have identified and characterized Glial cell line–Derived Neurotrophic Factor (GDNF) Family Receptor Alpha 1 (GFRA1) antibody–drug conjugates (ADC) using a cleavable valine-citrulline-MMAE (vcMMAE) linker-payload. RNAseq and IHC analysis confirmed the abundant expression of GFRA1 in luminal A breast cancer tissues, whereas minimal or no expression was observed in most normal tissues. Anti–GFRA-vcMMAE ADC internalized to the lysosomes and exhibited target-dependent killing of GFRA1-expressing cells both in vitro and in vivo. The ADCs using humanized anti-GFRA1 antibodies displayed robust therapeutic activity in clinically relevant cell line–derived (MCF7 and KPL-1) tumor xenograft models. The lead anti-GFRA1 ADC cross-reacts with rodent and cynomolgus monkey GFRA1 antigen and showed optimal pharmacokinetic properties in both species. These properties subsequently enabled a target-dependent toxicity study in rats. Anti-GFRA1 ADC is well tolerated in rats, as seen with other vcMMAE linker–payload based ADCs. Overall, these data suggest that anti–GFRA1-vcMMAE ADC may provide a targeted therapeutic opportunity for luminal A breast cancer patients. Mol Cancer Ther; 17(3); 638–49. ©2017 AACR.

Collaboration


Dive into the Rachana Ohri's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge