Antibody Therapy Section
Mitchell Ho, Ph.D.
Research
Dr. Ho's laboratory studies cell surface proteins in broad scientific fields of molecular and cellular biology including receptor/ligand interactions, signaling pathways, antibody/protein engineering, structure and computational biology, and functional genomics. We also develop new antibody technologies. Some of our research has direct clinical application.
Dissecting cell surface receptors in cancer: structure and function
Antibody-based therapy has shown promising efficacy in hematologic tumors. However, this approach has shown limitations in most solid cancers. One of the critical factors for treating solid tumors is the identification of cancer-specific targets. Our long-term research interests lie primarily in the biology of cell surface receptors including glypicans (GPC3 and GPC2) and mesothelin for establishing them as cancer targets.
Mesothelin is a target candidate in many solid tumors. To understand its biological function, we collaborated with Byungkook Lee (NCI) to make a structure model and experimentally identified the functional binding domain for MUC16/CA125 around Y318 (residues 296-359; named IAB) consisting of 64 amino acids at the N-terminus of cell surface mesothelin. Our work supports the role of mesothelin and MUC16/CA125 as functional partners in the tumor microenvironment and cancer development.
Heparan and chondroitin sulfate proteoglycans (HSPGs and CSPGs, respectively) are important modulators of signal transduction pathways during development and disease. HSPGs mainly consist of glycosylphosphatidylinositol (GPI)-anchored glypicans and transmembrane syndecans. Much of our work has focused on fundamental aspects of glypicans in connections to Wnt/Yap signaling and cancer cell growth. Our goal is to investigate glypicans as new targets in cancer.
We identified the Wnt functional binding sites on GPC3, one in the protein core and the other in heparan sulfate chains. First, we built a structure model of the GPC3/Wnt complex in collaboration with Byungkook Lee, then experimentally validated our model to determine the Wnt functional binding site on GPC3, providing evidence for GPC3 as a co-receptor for regulating Wnt. To explore the role of GPC3 in other signaling pathways, we conducted the original research that revealed the role of GPC3 in regulating Yap for liver cancer cell proliferation and discovered another signaling interaction between GPC3 and HGF in liver cancer. Recently, we identified FAT1, a potential cell surface receptor of Yap signaling in mammalian cells, was a novel GPC3 interacting protein in liver cancer cells. Furthermore, we found that our Wnt blocking single domain antibody (HN3) was capable of reaching the previously undescribed Wnt functional binding site in the N-lobe of the protein core of GPC3 and that F41 was the key residue in the hydrophobic groove for Wnt and HN3 binding. Additionally, we used our HS20 human antibody specific for the heparan sulfate chains of GPC3 to determine the Wnt binding motif which may function as a Wnt storage/transporter site on heparan sulfate chains. In collaboration with Jian Liu (University of North Carolina), we found that Wnt recognized a heparan sulfate structure containing IdoA2S and GlcNS6S, and that the 3-O-sulfation in GlcNS6S3S could enhance the binding of Wnt.
We started to apply what we learned from our GPC3 project to explore other glypicans in cancer. We found that GPC2 protein was expressed in nearly half of neuroblastoma cases and that GPC2 knockout inactivated Wnt/β-catenin signaling and reduced the expression of the target gene, N-Myc, an oncogenic driver of neuroblastoma tumorigenesis. Furthermore, we conducted the proof-of-concept study that showed CAR T cells and immunotoxins targeting GPC2 could inhibit neuroblastoma growth in mouse models. As a result, we reported GPC2 to be a new therapeutic target in neuroblastoma.
Engineering antibody therapeutics for treating cancer: immunotoxins and CAR T cells
To overcome the limitations of antibody-based immunotherapy for solid tumors, we used multiple antibody technologies to generate large panels of human and xenogeneic antibodies and utilized multiple effector mechanisms to improve the anti-tumor activity of our tumor antigen-specific monoclonal antibodies. We have developed multiple strategies with different mechanisms of actions, such as immunotoxins, ADCs and CAR T cells. Our research program focuses on the development and implementation of antibody-based immunotherapeutic strategies for the treatment of solid tumors, including hepatocellular carcinoma (HCC), pancreatic cancer, neuroblastoma and mesothelioma. The strategies developed in our program may be applicable to other solid tumors.
We generated antibodies (HN3 and YP7) that bind glypican-3 (GPC3) on liver cancer cells. HN3 is a human single domain antibody isolated using phage display technology, which recognizes a cryptic Wnt binding site in the N-lobe of GPC3. YP7 is a monoclonal antibody isolated using mouse hybridoma technology, which binds an epitope at the C-lobe of GPC3 close to cell surface. Using these antibodies, we generated immunotoxins and ADCs to target GPC3-expressing cancers. We constructed a novel immunotoxin that can produce tumor regression via a dual mechanism of action. These immunotoxins inactivate cancer signaling via the antibody domain and inhibit protein synthesis via the toxin portion. This dual mechanism of action may be applicable to other antibody-toxin/drug conjugates for better anti-tumor efficacy. Immunogenicity and a short serum half-life may limit the ability of immunotoxins to transition to the clinic. To address these concerns, we engineered HN3-based immunotoxins in collaboration with Ira Pastan (NCI) to use various deimmunized Pseudomonas exotoxin (PE) domains and added an albumin binding single domain. We also designed CAR T cells using our GPC3 antibodies and demonstrated that humanized YP7 (hYP7) antibody derived CAR T cells had potent and persistent anti-tumor activity in orthotopic liver cancer mouse models. This was accomplished by using functional genomics sequencing and single cell-based T cell analysis.
Mesothelin is highly expressed in mesothelioma and various solid tumors. We previously isolated HN1, a human monoclonal antibody that disrupts the mesothelin-MUC16 interaction. Our recent research has been focused on the targeting of the membrane proximal region of mesothelin for enhanced anti-tumor activities. As a result, we isolated YP218, a unique rabbit monoclonal antibody targeting the C-terminal portion of mesothelin close to the tumor cell surface.
Developing antibody technology: single domain antibodies
We have developed a variety of antibody technologies to produce therapeutic antibodies. These include cell-based screening methodology for isolating antibodies using rabbit and mouse hybridomas, humanization of rabbit and mouse antibodies, and construction of phage display libraries for antibody discovery. Our antibody technologies, such as single domain antibody libraries, can be used to identify therapeutic and diagnostic antibodies for human diseases including cancer, infectious disease and neurological disease.
Single domain antibodies (also commonly called nanobodies) are known to bind restricted epitopes that may be inaccessible to conventional antibodies. To make a large single domain phage library, we established a method based on PCR-Extension Assembly and Self-Ligation (named ‘EASeL’) to construct a VNAR single domain antibody library from six nurse sharks (Ginglymostoma cirratum) in collaboration with Martin Flajnik (University of Maryland). We conducted next-generation sequencing analysis of 1.2 million shark VNAR single domains and showed that our shark single domain library is highly diverse. In addition, we applied the EASeL method to construct VHH single domain phage libraries from 20 camels (Camelus dromedaries). Single domain antibodies isolated from our phage libraries can have a high affinity (KD = 1 nM or less) for their tumor or viral antigens.
Antibody engineering is typically carried out by displaying antibody fragments on the surface of microorganisms (e.g. phage, bacteria and yeast). We established a new antibody engineering method known as 'mammalian cell display' that is adapted from yeast cell display. Using this approach, antibody fragments are expressed on human HEK-293 cells, and high affinity antigen binders are isolated from a combinatory library via flow cytometry.
Drug resistance is an important component of tumor biology that requires a complex cellular environment for study. We have established ex vivo tumor spheroid models using cell lines and primary patient cells in collaboration with Shuichi Takayama (University of Michigan) and V. Courtney Broaddus (UCSF). This allows us to study the molecular mechanisms of antibody drug resistance in a physiologically relevant cellular model. We have also used microarrays to profile gene expression in both spheroids and monolayers to identify new targets specific to the 3D biological structure of cancer.
Teaching Interests
BIOC301/302 - Biochemistry I/II
Team
Covers

GPC3-targeted CAR T cells for liver cancer therapy
Background and Aims: Glypican 3 (GPC3) is an oncofetal antigen involved in Wnt-dependent cell proliferation that is highly expressed in hepatocellular carcinoma (HCC). We investigated whether the functions of chimeric antigen receptors (CARs) that target GPC3 are affected by their antibody- binding properties.
Methods: We collected peripheral blood mononuclear cells from healthy donors and patients with HCC and used them to create CAR T cells, based on the humanized YP7 (hYP7) and HN3 antibodies, which have high affinities for the C-lobe and N-lobe of GPC3, respectively. NOD/SCID/IL- 2Rgcnull (NSG) mice were given intraperitoneal injections of luciferase-expressing (Luc) Hep3B or HepG2 cells and after xenograft tumors formed, mice were given injections of saline or untransduced T cells (mock control), or CAR (HN3) T cells or CAR (hYP7) T cells. In other NOD/SCID/IL-2Rgcnull (NSG) mice, HepG2-Luc or Hep3B-Luc cells were injected into liver, and after orthotopic tumors formed, mice were given 1 injection of CAR (hYP7) T cells or CD19 CAR T cells (control). We developed droplet digital polymerase chain reaction and genome sequencing methods to analyze persistent CAR T cells in mice.
Results: Injections of CAR (hYP7) T cells eliminated tumors in 66% of mice by week 3, whereas CAR (HN3) T cells did not reduce tumor burden. Mice given CAR (hYP7) T cells remained tumor free after re-challenge with additional Hep3B cells. The CAR T cells induced perforin- and granzyme-mediated apoptosis and reduced levels of active b-catenin in HCC cells. Mice injected with CAR (hYP7) T cells had persistent expansion of T cells and subsets of polyfunctional CAR T cells via antigen- induced selection. These T cells were observed in the tumor microenvironment and spleen for up to 7 weeks after CAR T- cell administration. Integration sites in pre-infusion CAR (HN3) and CAR (hYP7) T cells were randomly distributed, whereas integration into NUPL1 was detected in 3.9% of CAR (hYP7) T cells 5 weeks after injection into tumor-bearing mice and 18.1% of CAR (hYP7) T cells at week 7. There was no common site of integration in CAR (HN3) or CD19 CAR T cells from tumor-bearing mice.
Conclusions: In mice with xenograft or orthoptic liver tumors, CAR (hYP7) T cells eliminate GPC3- positive HCC cells, possibly by inducing perforin- and granzyme-mediated apoptosis or reducing Wnt signaling in tumor cells. GPC3-targeted CAR T cells might be developed for treatment of patients with HCC.
Persistent Polyfunctional Chimeric Antigen Receptor T Cells That Target Glypican 3 Eliminate Orthotopic Hepatocellular Carcinomas in Mice. Li D, Li N, Zhang YF, Fu H, Feng M, Schneider D, Su L, Wu X, Zhou J, Mackay S, Kramer J, Duan Z, Yang H, Kolluri A, Hummer AM, Torres MB, Zhu H, Hall MD, Luo X, Chen J, Wang Q, Abate-Daga D, Dropulic B, Hewitt SM, Orentas RJ, Greten TF, Ho M. Gastroenterology. 2020 Jun;158(8):2250-2265.e20.

Engineered immunotoxin targeting GPC3 promotes liver cancer regression in mice
Background and Aims: Treatment of hepatocellular carcinomas using our glypican‐3 (GPC3)‐targeting human nanobody (HN3) immunotoxins causes potent tumor regression by blocking protein synthesis and down‐regulating the Wnt signaling pathway. However, immunogenicity and a short serum half‐life may limit the ability of immunotoxins to transition to the clinic.
Approach and Results: To address these concerns, we engineered HN3‐based immunotoxins to contain various deimmunized Pseudomonas exotoxin (PE) domains. This included HN3‐T20, which was modified to remove T‐cell epitopes and contains a PE domain II truncation. We compared them to our previously reported B‐cell deimmunized immunotoxin (HN3‐mPE24) and our original HN3‐immunotoxin with a wild‐type PE domain (HN3‐PE38). All of our immunotoxins displayed high affinity to human GPC3, with HN3‐T20 having a KD value of 7.4 nM. HN3‐T20 retained 73% enzymatic activity when compared with the wild‐type immunotoxin in an adenosine diphosphate–ribosylation assay. Interestingly, a real‐time cell growth inhibition assay demonstrated that a single dose of HN3‐T20 at 62.5 ng/mL (1.6 nM) was capable of inhibiting nearly all cell proliferation during the 10‐day experiment. To enhance HN3‐T20’s serum retention, we tested the effect of adding a streptococcal albumin‐binding domain (ABD) and a llama single‐domain antibody fragment specific for mouse and human serum albumin. For the detection of immunotoxin in mouse serum, we developed a highly sensitive enzyme‐linked immunosorbent assay and found that HN3‐ABD‐T20 had a 45‐fold higher serum half‐life than HN3‐T20 (326 minutes vs. 7.3 minutes); consequently, addition of an ABD resulted in HN3‐ABD‐T20–mediated tumor regression at 1 mg/kg.
Conclusion: These data indicate that ABD‐containing deimmunized HN3‐T20 immunotoxins are high‐potency therapeutics ready to be evaluated in clinical trials for the treatment of liver cancer.
Engineered Anti-GPC3 Immunotoxin, HN3-ABD-T20, Produces Regression in Mouse Liver Cancer Xenografts Through Prolonged Serum Retention. Fleming BD, Urban DJ, Hall MD, Longerich T, Greten TF, Pastan I, Ho M. Hepatology. 2020 May;71(5):1696-1711.

A Shark Single Domain Antibody Library
The laboratory of Mitchell Ho at the National Cancer Institute has established a large phage-displayed VNAR single domain antibody library from nurse sharks. The shark single domain library provides a new platform for selecting therapeutic antibodies. For additional information, see Antibody Therapeutics 2019 Jan;2(1):1-11.
Construction and next-generation sequencing analysis of a large phage-displayed VNAR single-domain antibody library from six naïve nurse sharks. Feng M, Bian H, Wu X, Fu T, Fu Y, Hong J, Fleming BD, Flajnik MF, Ho M. Antibody Therapeutics 2019 Jan;2(1):1-11.

Searching for Magic Bullets
Single domain antibodies have emerged as a new class of therapeutic molecules in antibody engineering. A single domain antibody (shark VNAR), in contrast to a conventional antibody (Fv), is capable of binding a buried functional site consisting of the enzyme’s substrate binding pocket. For additional information, see Antibody Therapeutics 2018 Jun;1(1):1-5.
Inaugural Editorial: Searching for Magic Bullets. Ho M. Antibody Therapeutics 2018 Jun;1(1):1-5.

A novel anti-GPC3 monoclonal antibody (YP7)
Glypican-3 (GPC3) is an emerging therapeutic target in hepatoma. A novel anti-GPC3 monoclonal antibody (YP7) has been generated through a combination of peptide immunization and high-throughput flow cytometry screening. YP7 binds cell-surface-associated GPC3 with high affinity and exhibits significant hepatoma xenograft growth inhibition in nude mice. The new antibody may have potential for hepatoma therapy. For additional information, see MAbs. 2012 Sep-Oct;4(5):592-9.
High-affinity monoclonal antibodies to cell surface tumor antigen glypican-3 generated through a combination of peptide immunization and flow cytometry screening. MAbs. 2012 Sep-Oct;4(5):592-9.
Lab Life

Ho lab (2022)

Ho lab (2021)

Ho lab (2020)

Ho lab (2019)

Ho lab (2018)

Ho lab (2017)

Ho lab (2016)

Ho lab (2015)

Ho lab (2014)

Ho lab (2013)

Ho lab (2012)
Ho lab (2011)

Ho lab (2010)

Ho lab (2009)

Ho lab (2008)