Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
1.
MAbs ; 15(1): 2230618, 2023.
Article in English | MEDLINE | ID: mdl-37408311

ABSTRACT

The commentary by Colombo and Rich recently published in Cancer Cell provides a timely and comprehensive review of the clinical maximum tolerated doses (MTDs) of antibody-drug conjugates (ADCs) and their corresponding small molecules/chemotherapies. The authors identified similarities between their MTDs and therefore question the historic assumptions made for ADCs, namely, that they increase the MTDs of their corresponding cytotoxic molecules. However, the authors did not address the superior anti-tumor responses of ADCs compared to their corresponding chemotherapies, as reported in clinical trials. In this point of view, we propose a revised model wherein the anti-tumor activities of ADCs and consequently their therapeutic indexes (TIs) are not solely associated with changes not only in their MTDs but also in their minimal effective doses (MEDs). In addition, when using an exposure-based TI calculation method, the superior anti-tumor activities of ADCs relative to their corresponding chemotherapy can readily be explained. We discussed the clinical and preclinical data in support of lower MEDs of ADCs and generated a revised graph illustrating the TI improvements of ADCs vs chemotherapy more accurately. We believe that our revised model can provide a blueprint for future improvements in protein engineering and chemical engineering of toxins to further advance ADC research and development.


Subject(s)
Antineoplastic Agents , Immunoconjugates , Neoplasms , Humans , Immunoconjugates/therapeutic use , Immunoconjugates/chemistry , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/chemistry , Neoplasms/drug therapy , Protein Engineering , Xenograft Model Antitumor Assays
2.
Front Oncol ; 12: 1027548, 2022.
Article in English | MEDLINE | ID: mdl-36338746

ABSTRACT

pHLA complexes represent the largest class of cell surface markers on cancer cells, making them attractive for targeted cancer therapies. Adoptive cell therapies expressing TCRs that recognize tumor specific pHLAs take advantage of the unique selectivity and avidity of TCR: pHLA interactions. More recently, additional protein binding domains binding to pHLAs, known as TCR mimics (TCRm), were developed for tumor targeting of high potency therapeutic modalities, including bispecifics, ADCs, CAR T and -NK cells. TCRm compounds take advantage of the exquisite tumor specificity of certain pHLA targets, including cell lineage commitment markers and cancer testis antigens (CTAs). To achieve meaningful anti-tumor responses, it is critical that TCRm compounds integrate both, high target binding affinities and a high degree of target specificity. In this review, we describe the most advanced approaches to achieve both criteria, including affinity- and specificity engineering of TCRs, antibodies and alternative protein scaffolds. We also discuss the status of current TCRm based therapeutics developed in the clinic, key challenges, and emerging trends to improve treatment options for cancer patients treated with TCRm based therapeutics in Oncology.

3.
Mol Cancer Ther ; 21(9): 1462-1472, 2022 09 06.
Article in English | MEDLINE | ID: mdl-35793468

ABSTRACT

Extra domain B splice variant of fibronectin (EDB+FN) is an extracellular matrix protein (ECM) deposited by tumor-associated fibroblasts, and is associated with tumor growth, angiogenesis, and invasion. We hypothesized that EDB+FN is a safe and abundant target for therapeutic intervention with an antibody-drug conjugate (ADC). We describe the generation, pharmacology, mechanism of action, and safety profile of an ADC specific for EDB+FN (EDB-ADC). EDB+FN is broadly expressed in the stroma of pancreatic, non-small cell lung (NSCLC), breast, ovarian, head and neck cancers, whereas restricted in normal tissues. In patient-derived xenograft (PDX), cell-line xenograft (CLX), and mouse syngeneic tumor models, EDB-ADC, conjugated to auristatin Aur0101 through site-specific technology, demonstrated potent antitumor growth inhibition. Increased phospho-histone H3, a pharmacodynamic biomarker of response, was observed in tumor cells distal to the target site of tumor ECM after EDB-ADC treatment. EDB-ADC potentiated infiltration of immune cells, including CD3+ T lymphocytes into the tumor, providing rationale for the combination of EDB-ADC with immune checkpoint therapy. EDB-ADC and anti-PD-L1 combination in a syngeneic breast tumor model led to enhanced antitumor activity with sustained tumor regressions. In nonclinical safety studies in nonhuman primates, EDB-ADC had a well-tolerated safety profile without signs of either on-target toxicity or the off-target effects typically observed with ADCs that are conjugated through conventional conjugation methods. These data highlight the potential for EDB-ADC to specifically target the tumor microenvironment, provide robust therapeutic benefits against multiple tumor types, and enhance activity antitumor in combination with checkpoint blockade.


Subject(s)
Breast Neoplasms , Immunoconjugates , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Fibronectins/metabolism , Humans , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Mice , Neovascularization, Pathologic/metabolism , Tumor Microenvironment , Xenograft Model Antitumor Assays
4.
Cell Rep Med ; 2(5): 100279, 2021 05 18.
Article in English | MEDLINE | ID: mdl-34095881

ABSTRACT

Aberrant NOTCH3 signaling and overexpression is oncogenic, associated with cancer stem cells and drug resistance, yet therapeutic targeting remains elusive. Here, we develop NOTCH3-targeted antibody drug conjugates (NOTCH3-ADCs) by bioconjugation of an auristatin microtubule inhibitor through a protease cleavable linker to two antibodies with differential abilities to inhibit signaling. The signaling inhibitory antibody rapidly induces ligand-independent receptor clustering and internalization through both caveolin and clathrin-mediated pathways. The non-inhibitory antibody also efficiently endocytoses via clathrin without inducing receptor clustering but with slower lysosomal co-localization kinetics. In addition, DLL4 ligand binding to the NOTCH3 receptor mediates transendocytosis of NOTCH3-ADCs into ligand-expressing cells. NOTCH3-ADCs internalize into receptor and ligand cells independent of signaling and induce cell death in both cell types representing an atypical mechanism of ADC cytotoxicity. Treatment of xenografts with NOTCH3-ADCs leads to sustained tumor regressions, outperforms standard-of-care chemotherapy, and allows targeting of tumors that overexpress NOTCH3 independent of signaling inhibition.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Immunoconjugates/pharmacology , Receptor, Notch3/metabolism , Cell Line, Tumor/drug effects , Humans , Immunoconjugates/metabolism , Oncogenes/drug effects , Receptor, Notch3/immunology , Receptors, Notch/antagonists & inhibitors , Xenograft Model Antitumor Assays
5.
Clin Cancer Res ; 27(2): 622-631, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33148666

ABSTRACT

PURPOSE: Mortality due to acute myeloid leukemia (AML) remains high, and the management of relapsed or refractory AML continues to be therapeutically challenging. The reapproval of Mylotarg, an anti-CD33-calicheamicin antibody-drug conjugate (ADC), has provided a proof of concept for an ADC-based therapeutic for AML. Several other ADCs have since entered clinical development of AML, but have met with limited success. We sought to develop a next-generation ADC for AML with a wide therapeutic index (TI) that overcomes the shortcomings of previous generations of ADCs. EXPERIMENTAL DESIGN: We compared the TI of our novel CD33-targeted ADC platform with other currently available CD33-targeted ADCs in preclinical models of AML. Next, using this next-generation ADC platform, we performed a head-to-head comparison of two attractive AML antigens, CD33 and CD123. RESULTS: Our novel ADC platform offered improved safety and TI when compared with certain currently available ADC platforms in preclinical models of AML. Differentiation between the CD33- and CD123-targeted ADCs was observed in safety studies conducted in cynomolgus monkeys. The CD33-targeted ADC produced severe hematologic toxicity, whereas minimal hematologic toxicity was observed with the CD123-targeted ADC at the same doses and exposures. The improved toxicity profile of an ADC targeting CD123 over CD33 was consistent with the more restricted expression of CD123 in normal tissues. CONCLUSIONS: We optimized all components of ADC design (i.e., leukemia antigen, antibody, and linker-payload) to develop an ADC that has the potential to translate into an effective new therapy against AML.


Subject(s)
Gemtuzumab/therapeutic use , Immunoconjugates/therapeutic use , Interleukin-3 Receptor alpha Subunit/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Sialic Acid Binding Ig-like Lectin 3/antagonists & inhibitors , Animals , Antineoplastic Agents, Immunological/immunology , Antineoplastic Agents, Immunological/pharmacokinetics , Antineoplastic Agents, Immunological/therapeutic use , Area Under Curve , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Gemtuzumab/immunology , Gemtuzumab/pharmacokinetics , HL-60 Cells , Humans , Immunoconjugates/immunology , Immunoconjugates/pharmacokinetics , Interleukin-3 Receptor alpha Subunit/immunology , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/metabolism , Macaca fascicularis , Mice , Sialic Acid Binding Ig-like Lectin 3/immunology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays/methods
6.
Mol Cancer Ther ; 19(10): 2068-2078, 2020 10.
Article in English | MEDLINE | ID: mdl-32747418

ABSTRACT

The approval of ado-trastuzumab emtansine (T-DM1) in HER2+ metastatic breast cancer validated HER2 as a target for HER2-specific antibody-drug conjugates (ADC). Despite its demonstrated clinical efficacy, certain inherent properties within T-DM1 hamper this compound from achieving the full potential of targeting HER2-expressing solid tumors with ADCs. Here, we detail the discovery of PF-06804103, an anti-HER2 ADC designed to have a widened therapeutic window compared with T-DM1. We utilized an empirical conjugation site screening campaign to identify the engineered ĸkK183C and K290C residues as those that maximized in vivo ADC stability, efficacy, and safety for a four drug-antibody ratio (DAR) ADC with this linker-payload combination. PF-06804103 incorporates the following novel design elements: (i) a new auristatin payload with optimized pharmacodynamic properties, (ii) a cleavable linker for optimized payload release and enhanced antitumor efficacy, and (iii) an engineered cysteine site-specific conjugation approach that overcomes the traditional safety liabilities of conventional conjugates and generates a homogenous drug product with a DAR of 4. PF-06804103 shows (i) an enhanced efficacy against low HER2-expressing breast, gastric, and lung tumor models, (ii) overcomes in vitro- and in vivo-acquired T-DM1 resistance, and (iii) an improved safety profile by enhancing ADC stability, pharmacokinetic parameters, and reducing off-target toxicities. Herein, we showcase our platform approach in optimizing ADC design, resulting in the generation of the anti-HER2 ADC, PF-06804103. The design elements of identifying novel sites of conjugation employed in this study serve as a platform for developing optimized ADCs against other tumor-specific targets.


Subject(s)
Breast Neoplasms/drug therapy , Immunoconjugates/therapeutic use , Lung Neoplasms/drug therapy , Stomach Neoplasms/drug therapy , Animals , Breast Neoplasms/pathology , Female , Humans , Immunoconjugates/pharmacology , Lung Neoplasms/pathology , Mice , Mice, Nude , Stomach Neoplasms/pathology
7.
Trends Cancer ; 6(4): 299-318, 2020 04.
Article in English | MEDLINE | ID: mdl-32209445

ABSTRACT

The ideal cancer target antigen (Ag) is expressed at high copy numbers on neoplastic cells, absent on normal tissues, and contributes to the survival of cancer cells. Despite significant investments in the identification of cell surface Ags, there is a paucity of targets that meet such ideal cancer target criteria. Recent clinical trials in patients with cancer treated with immune checkpoint inhibitors (ICIs) indicate that cluster of differentiation (CD)8+ T cells, by means of their T cell receptors (TCRs) recognizing intracellular targets presented as peptides in the context of human leukocyte antigen (peptide-human leukocyte antigen complex; pHLA) molecules on tumor cells, can mediate deep and long-lasting antitumor responses in patients with solid tumors. Therefore, pHLA-target Ags may represent the long sought-after, ideal targets for solid tumor targeting by high-potency oncology compounds.


Subject(s)
Antigens, Neoplasm/metabolism , Antineoplastic Agents, Immunological/pharmacology , Drug Discovery/methods , Neoplasms/drug therapy , Receptors, Antigen, T-Cell/metabolism , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Antineoplastic Agents, Immunological/therapeutic use , Cell Line, Tumor , Computer Simulation , Cross Reactions/immunology , Epitope Mapping/methods , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , HLA Antigens/genetics , HLA Antigens/immunology , HLA Antigens/metabolism , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Ligands , Neoplasms/immunology , Neoplasms/pathology , Peptide Library , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
8.
Biochem Pharmacol ; 168: 275-284, 2019 10.
Article in English | MEDLINE | ID: mdl-31323186

ABSTRACT

High-potency oncology compounds such as antibody- drug conjugates, T cell redirecting, and CAR-T cell therapies have provided transformational responses in patients with liquid tumors. However, they delivered only limited benefit to solid tumor patients due to the frequent onset of dose limiting toxicities in normal tissues. Such on-target, off-tumor toxicities are caused by recognition of targets present at low-levels on normal tissues. The apparent imbalance between the rapid development of high-potency therapeutic modalities and the slow progress in identification of cleaner targets is illustrated by the fact that most high-potency compounds currently developed in the clinic target cell surface antigens identified over 20 years ago. Therefore, identification of novel, truly tumor-specific targets is critical for the future success of high-potency oncology compounds in solid tumors. One of the most promising approaches to overcome the limitations of targeting cell surface antigens are intracellular targets. The renewed interest in this class of targets is due to the success of immune checkpoint inhibitors, which mediate their anti-tumor responses by activation of cytotoxic T cells recognizing peptide fragments of intracellular targets presented by human leukocyte antigens (HLAs) on the surface of tumor cells. Importantly, many intracellular targets belong to the class of tumor specific antigens (TSAs), which lack presentation on normal tissues. In this report we review the main classes of tumor specific antigens, including viral, neoantigens and shared self-antigens as well as tumor associated antigens (TAAs) and their relevance for therapeutic targeting of solid tumors by high-potency therapeutic modalities.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Delivery Systems/methods , Intracellular Fluid/drug effects , Neoplasms/drug therapy , Animals , Antigens, Neoplasm/drug effects , Antigens, Neoplasm/metabolism , Antineoplastic Agents/metabolism , Drug Delivery Systems/trends , Humans , Intracellular Fluid/metabolism , Neoplasms/metabolism , Treatment Outcome
9.
Mol Cancer Ther ; 17(1): 243-253, 2018 01.
Article in English | MEDLINE | ID: mdl-29054985

ABSTRACT

Trastuzumab emtansine (T-DM1) is an antibody-drug conjugate (ADC) that has demonstrated clinical benefit for patients with HER2+ metastatic breast cancer; however, its clinical activity is limited by inherent or acquired drug resistance. The molecular mechanisms that drive clinical resistance to T-DM1, especially in HER2+ tumors, are not well understood. We used HER2+ cell lines to develop models of T-DM1 resistance using a cyclical dosing schema in which cells received T-DM1 in an "on-off" routine until a T-DM1-resistant population was generated. T-DM1-resistant N87 cells (N87-TM) were cross-resistant to a panel of trastuzumab-ADCs (T-ADCs) with non-cleavable-linked auristatins. N87-TM cells do not have a decrease in HER2 protein levels or an increase in drug transporter protein (e.g., MDR1) expression compared with parental N87 cells. Intriguingly, T-ADCs using auristatin payloads attached via an enzymatically cleavable linker overcome T-DM1 resistance in N87-TM cells. Importantly, N87-TM cells implanted into athymic mice formed T-DM1 refractory tumors that remain sensitive to T-ADCs with cleavable-linked auristatin payloads. Comparative proteomic profiling suggested enrichment in proteins that mediate caveolae formation and endocytosis in the N87-TM cells. Indeed, N87-TM cells internalize T-ADCs into intracellular caveolin-1 (CAV1)-positive puncta and alter their trafficking to the lysosome compared with N87 cells. T-DM1 colocalization into intracellular CAV1-positive puncta correlated with reduced response to T-DM1 in a panel of HER2+ cell lines. Together, these data suggest that caveolae-mediated endocytosis of T-DM1 may serve as a novel predictive biomarker for patient response to T-DM1. Mol Cancer Ther; 17(1); 243-53. ©2017 AACR.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Endocytosis/drug effects , Trastuzumab/therapeutic use , Animals , Antineoplastic Agents, Immunological/pharmacology , Caveolae , Drug Resistance, Neoplasm , Female , Humans , Male , Mice , Trastuzumab/pharmacology
10.
Cancer Immunol Immunother ; 67(2): 247-259, 2018 02.
Article in English | MEDLINE | ID: mdl-29067496

ABSTRACT

Strong evidence exists supporting the important role T cells play in the immune response against tumors. Still, the ability to initiate tumor-specific immune responses remains a challenge. Recent clinical trials suggest that bispecific antibody-mediated retargeted T cells are a promising therapeutic approach to eliminate hematopoietic tumors. However, this approach has not been validated in solid tumors. PF-06671008 is a dual-affinity retargeting (DART®)-bispecific protein engineered with enhanced pharmacokinetic properties to extend in vivo half-life, and designed to engage and activate endogenous polyclonal T cell populations via the CD3 complex in the presence of solid tumors expressing P-cadherin. This bispecific molecule elicited potent P-cadherin expression-dependent cytotoxic T cell activity across a range of tumor indications in vitro, and in vivo in tumor-bearing mice. Regression of established tumors in vivo was observed in both cell line and patient-derived xenograft models engrafted with circulating human T lymphocytes. Measurement of in vivo pharmacodynamic markers demonstrates PF-06671008-mediated T cell activation, infiltration and killing as the mechanism of tumor inhibition.


Subject(s)
Antibodies, Bispecific/immunology , Antibodies, Bispecific/pharmacology , Cadherins/immunology , Immunotherapy/methods , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes/immunology , Animals , CD3 Complex/immunology , Cell Line, Tumor , Cricetinae , Cricetulus , Female , HCT116 Cells , HT29 Cells , Humans , Mice , Xenograft Model Antitumor Assays
11.
PLoS One ; 12(7): e0179561, 2017.
Article in English | MEDLINE | ID: mdl-28727782

ABSTRACT

The fetal oncogene 5T4 is a cell surface protein, with overexpression observed in a variety of cancers as compared to normal adult tissue. The ability to select patients with tumors that express high levels of 5T4 may enrich a clinical trial cohort with patients most likely to respond to 5T4 targeted therapy. To that end, we developed assays to measure 5T4 in both tumors and in circulating tumor cells (CTCs). We identified the presence of 5T4 in both adenocarcinoma and squamous cell carcinoma of lung, in all clinical stages and grades of disease. CTCs were identified in peripheral blood from the majority of patients with NSCLC, and 5T4 was detectable in most samples. Although 5T4 was present in both CTCs and tumors in most patients, there was no concordance between relative amount in either sample type. Clinical response rates of patients treated with the therapies directed against 5T4 in early stage clinical trials, as determined by these assays, may provide important insights into the biology of 5T4 in tumors and the mechanisms of action of 5T4-targeting therapy.


Subject(s)
Adenocarcinoma/metabolism , Carcinoma, Squamous Cell/metabolism , Lung Neoplasms/metabolism , Membrane Glycoproteins/metabolism , Neoplastic Cells, Circulating/metabolism , Adenocarcinoma/pathology , Animals , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Female , Heterografts , Humans , Immunohistochemistry , Lung Neoplasms/pathology , Male , Mice , Neoplasm Transplantation , Neoplastic Cells, Circulating/pathology
12.
Clin Cancer Res ; 23(7): 1760-1770, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-27683177

ABSTRACT

Purpose: Adverse reactions reported in patients treated with antibody-calicheamicin conjugates such as gemtuzumab ozogamicin (Mylotarg) and inotuzumab ozogamicin include thrombocytopenia and sinusoidal obstruction syndrome (SOS). The objective of this experimental work was to investigate the mechanism for thrombocytopenia, characterize the liver injury, and identify potential safety biomarkers.Experimental Design: Cynomolgus monkeys were dosed intravenously at 6 mg/m2/dose once every 3 weeks with a nonbinding antibody-calicheamicin conjugate (PF-0259) containing the same linker-payload as gemtuzumab ozogamicin and inotuzumab ozogamicin. Monkeys were necropsied 48 hours after the first administration (day 3) or 3 weeks after the third administration (day 63).Results: PF-0259 induced acute thrombocytopenia (up to 86% platelet reduction) with nadirs on days 3 to 4. There was no indication of effects on megakaryocytes in bone marrow or activation of platelets in peripheral blood. Microscopic evaluation of liver from animals necropsied on day 3 demonstrated midzonal degeneration and loss of sinusoidal endothelial cells (SECs) associated with marked platelet accumulation in sinusoids. Liver histopathology on day 63 showed variable endothelial recovery and progression to a combination of sinusoidal capillarization and sinusoidal dilation/hepatocellular atrophy, consistent with early SOS. Among biomarkers evaluated, there were early and sustained increases in serum hyaluronic acid (HA) that correlated well with serum aspartate aminotransferase and liver microscopic changes, suggesting that HA may be a sensitive diagnostic marker of the liver microvascular injury.Conclusions: These data support the conclusion that target-independent damage to liver SECs may be responsible for acute thrombocytopenia (through platelet sequestration in liver sinusoids) and development of SOS. Clin Cancer Res; 23(7); 1760-70. ©2016 AACR.


Subject(s)
Aminoglycosides/administration & dosage , Antibodies, Monoclonal, Humanized/administration & dosage , Chemical and Drug Induced Liver Injury/pathology , Thrombocytopenia/pathology , Aminoglycosides/adverse effects , Aminoglycosides/chemistry , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized/adverse effects , Endothelial Cells/drug effects , Endothelial Cells/pathology , Enediynes/administration & dosage , Enediynes/chemistry , Gemtuzumab , Humans , Hyaluronic Acid/blood , Immunoconjugates/administration & dosage , Immunoconjugates/adverse effects , Inotuzumab Ozogamicin , Liver/drug effects , Macaca fascicularis , Neoplasms/drug therapy , Neoplasms/pathology , Thrombocytopenia/chemically induced
13.
14.
Mol Cancer Ther ; 15(12): 2825-2834, 2016 12.
Article in English | MEDLINE | ID: mdl-27780876

ABSTRACT

Drug resistance limits the effectiveness of cancer therapies. Despite attempts to develop curative anticancer treatments, tumors evolve evasive mechanisms limiting durable responses. Hence, diverse therapies are used to attack cancer, including cytotoxic and targeted agents. Antibody-drug conjugates (ADC) are biotherapeutics designed to deliver potent cytotoxins to cancer cells via tumor-specific antigens. Little is known about the clinical manifestations of drug resistance to this class of therapy; however, recent preclinical studies reveal potential mechanisms of resistance. Because ADCs are a combination of antibody and small molecule cytotoxin, multifactorial modes of resistance are emerging that are inherent to the structure and function of the ADC. Decreased cell-surface antigen reduces antibody binding, whereas elevated drug transporters such as MDR1 and MRP1 reduce effectiveness of the payload. Inherent to the uniqueness of the ADC, other novel resistance mechanisms are emerging, including altered antibody trafficking, ADC processing, and intracellular drug release. Most importantly, the modular nature of the ADC allows components to be switched and replaced, enabling development of second-generation ADCs that overcome acquired resistance. This review is intended to highlight recent progress in our understanding of ADC resistance, including approaches to create preclinical ADC-refractory models and to characterize their emerging mechanisms of resistance. Mol Cancer Ther; 15(12); 2825-34. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Immunoconjugates/pharmacology , Neoplasms/drug therapy , Animals , Antibodies, Monoclonal/administration & dosage , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Antineoplastic Agents/therapeutic use , Cytotoxins/administration & dosage , Drug Design , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/immunology , Gene Expression Regulation , Humans , Immunoconjugates/therapeutic use , Molecular Targeted Therapy , Signal Transduction
15.
Biochem Pharmacol ; 102: 1-6, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26686577

ABSTRACT

Blockade of immune-checkpoints has emerged as one of the most promising approaches to improve the durability of anti-tumor responses in cancer patients. However, the fraction of patients experiencing durable responses to single agent immune checkpoint inhibitor treatment remains limited. Recent clinical reports suggest that patients responding best to checkpoint blockade therapies display higher levels of CD8(+) T-cells in the tumor prior to treatment. Therefore, combination treatments of immune-checkpoint inhibitors with compounds that increase the number of tumor infiltrating CD8(+) T cells may expand the therapeutic benefit of immuno-oncology (IO) drugs. Immunogenic cell death (ICD) of tumor cells is induced by certain classes of cytotoxic compounds and represents a potent stimulator of effector T-cell recruitment to tumors. In addition, several cytotoxics directly stimulate dendritic cell activation and maturation, resulting in improved anti-tumor immune responses when combined with IO compounds. Among them, several cytotoxic agents are currently utilized as payloads for antibody-drug conjugates (ADCs). Therefore, identification of optimal combination regimens between ADC- and IO compounds holds strong promise to overcome the current limitations of immune checkpoint inhibitors, by increasing the recruitment of CD8(+) effector T-cells to the tumor core. Here we review the emerging field of ADC/IO combination research, with a focus on how to optimally combine both modalities. The answer to this question may have a broader impact on oncology drug development, as synergistic activities between IO compounds and ADCs may increase the formation of tumor specific immunological memory, ultimately leading to durable responses in a larger fraction of cancer patients.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antineoplastic Agents/administration & dosage , Immunologic Factors/administration & dosage , Immunotherapy/methods , Neoplasms/therapy , Animals , Antibodies, Monoclonal/immunology , Antigens, Neoplasm/administration & dosage , Antigens, Neoplasm/immunology , Antineoplastic Agents/immunology , CD8-Positive T-Lymphocytes/immunology , Combined Modality Therapy/methods , Humans , Immunologic Factors/immunology , Neoplasms/immunology
16.
Antibodies (Basel) ; 5(1)2016 Mar 04.
Article in English | MEDLINE | ID: mdl-31557987

ABSTRACT

Bispecific antibodies offer a promising approach for the treatment of cancer but can be challenging to engineer and manufacture. Here we report the development of PF-06671008, an extended-half-life dual-affinity re-targeting (DART®) bispecific molecule against P-cadherin and CD3 that demonstrates antibody-like properties. Using phage display, we identified anti-P-cadherin single chain Fv (scFv) that were subsequently affinity-optimized to picomolar affinity using stringent phage selection strategies, resulting in low picomolar potency in cytotoxic T lymphocyte (CTL) killing assays in the DART format. The crystal structure of this disulfide-constrained diabody shows that it forms a novel compact structure with the two antigen binding sites separated from each other by approximately 30 Å and facing approximately 90° apart. We show here that introduction of the human Fc domain in PF-06671008 has produced a molecule with an extended half-life (-4.4 days in human FcRn knock-in mice), high stability (Tm1 > 68 °C), high expression (>1 g/L), and robust purification properties (highly pure heterodimer), all with minimal impact on potency. Finally, we demonstrate in vivo anti-tumor efficacy in a human colorectal/human peripheral blood mononuclear cell (PBMC) co-mix xenograft mouse model. These results suggest PF-06671008 is a promising new bispecific for the treatment of patients with solid tumors expressing P-cadherin.

17.
Clin Cancer Res ; 21(18): 4165-73, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26015513

ABSTRACT

PURPOSE: Triple-negative breast cancer (TNBC) and ovarian cancer each comprise heterogeneous tumors, for which current therapies have little clinical benefit. Novel therapies that target and eradicate tumor-initiating cells (TIC) are needed to significantly improve survival. EXPERIMENTAL DESIGN: A panel of well-annotated patient-derived xenografts (PDX) was established, and surface markers that enriched for TIC in specific tumor subtypes were empirically determined. The TICs were queried for overexpressed antigens, one of which was selected to be the target of an antibody-drug conjugate (ADC). The efficacy of the ADC was evaluated in 15 PDX models to generate hypotheses for patient stratification. RESULTS: We herein identified E-cadherin (CD324) as a surface antigen able to reproducibly enrich for TIC in well-annotated, low-passage TNBC and ovarian cancer PDXs. Gene expression analysis of TIC led to the identification of Ephrin-A4 (EFNA4) as a prospective therapeutic target. An ADC comprising a humanized anti-EFNA4 monoclonal antibody conjugated to the DNA-damaging agent calicheamicin achieved sustained tumor regressions in both TNBC and ovarian cancer PDX in vivo. Non-claudin low TNBC tumors exhibited higher expression and more robust responses than other breast cancer subtypes, suggesting a specific translational application for tumor subclassification. CONCLUSIONS: These findings demonstrate the potential of PF-06647263 (anti-EFNA4-ADC) as a first-in-class compound designed to eradicate TIC. The use of well-annotated PDX for drug discovery enabled the identification of a novel TIC target, pharmacologic evaluation of the compound, and translational studies to inform clinical development.


Subject(s)
Aminoglycosides/chemistry , Antibodies, Monoclonal, Murine-Derived/chemistry , Enediynes/chemistry , Ephrin-A4/chemistry , Ovarian Neoplasms/drug therapy , Triple Negative Breast Neoplasms/drug therapy , Animals , Antibodies, Monoclonal, Humanized/chemistry , Antigens, Neoplasm/chemistry , Cell Line, Tumor , DNA/chemistry , Drug Design , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/metabolism , Prospective Studies , Random Allocation , Treatment Outcome , Xenograft Model Antitumor Assays
18.
Mol Cancer Ther ; 14(4): 952-63, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25646013

ABSTRACT

Antibody-drug conjugates (ADC) are emerging as clinically effective therapy. We hypothesized that cancers treated with ADCs would acquire resistance mechanisms unique to immunoconjugate therapy and that changing ADC components may overcome resistance. Breast cancer cell lines were exposed to multiple cycles of anti-Her2 trastuzumab-maytansinoid ADC (TM-ADC) at IC80 concentrations followed by recovery. The resistant cells, 361-TM and JIMT1-TM, were characterized by cytotoxicity, proteomic, transcriptional, and other profiling. Approximately 250-fold resistance to TM-ADC developed in 361-TM cells, and cross-resistance was observed to other non-cleavable-linked ADCs. Strikingly, these 361-TM cells retained sensitivity to ADCs containing cleavable mcValCitPABC-linked auristatins. In JIMT1-TM cells, 16-fold resistance to TM-ADC developed, with cross-resistance to other trastuzumab-ADCs. Both 361-TM and JIMT1-TM cells showed minimal resistance to unconjugated mertansine (DM1) and other chemotherapeutics. Proteomics and immunoblots detected increased ABCC1 (MRP1) drug efflux protein in 361-TM cells, and decreased Her2 (ErbB2) in JIMT1-TM cells. Proteomics also showed alterations in various pathways upon chronic exposure to the drug in both cell models. Tumors derived from 361-TM cells grew in mice and were refractory to TM-ADC compared with parental cells. Hence, acquired resistance to trastuzumab-maytansinoid ADC was generated in cultured cancer cells by chronic drug treatment, and either increased ABCC1 protein or reduced Her2 antigen were primary mediators of resistance. These ADC-resistant cell models retain sensitivity to other ADCs or standard-of-care chemotherapeutics, suggesting that alternate therapies may overcome acquired ADC resistance. Mol Cancer Ther; 14(4); 952-63. ©2015 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Immunoconjugates/pharmacology , Trastuzumab/pharmacology , Animals , Antigens, Surface/genetics , Antigens, Surface/metabolism , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Female , Gene Expression Profiling , Humans , Immunoconjugates/administration & dosage , Inhibitory Concentration 50 , Mice , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , Protein Transport , Proteome , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Signal Transduction , Transcriptome , Trastuzumab/administration & dosage , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
19.
Mol Immunol ; 67(2 Pt A): 107-16, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25304309

ABSTRACT

Calicheamicin is a DNA-damaging agent that, following intracellular activation, binds to DNA in the minor groove and introduces double-strand DNA breaks, leading to G2/M arrest and subsequent cell death. Importantly, the mechanism of action of calicheamicin is fundamentally different from the tubulin-binding class of cytotoxics targeting the mitotic spindle, which represent the most common class of payloads for antibody-drug conjugates (ADCs) currently undergoing clinical development. Spindle poisons that target tubulin, including auristatins and maytansines, are most effective against rapidly proliferating cells. In contrast, calicheamicin induces DNA double-strand breaks and apoptosis independent of cell cycle progression. Such properties may be advantageous when targeting malignant cells that are not markedly different in their proliferation status compared to normal cells. Here we review calicheamicin conjugates, with a particular focus on the preclinical- and clinical development of inotuzumab ozogamicin, targeting the CD22 antigen expressed on a large variety of hematologic malignancies. In pre-clinical experiments, inotuzumab ozogamicin potently induced tumor regressions in models of non-Hodgkin's lymphoma (NHL), either alone or in combination with the anti-CD20 antibody Rituximab. Promising anti-tumor responses were observed in early stage clinical trials, where inotuzumab ozogamicin was administered either as single agent or in combination with Rituximab. Consistent with the cell cycle independent mechanism of action of the calicheamicin payload, high rates of complete responses were observed in less aggressive forms of lymphomas, including follicular lymphoma (FL) and relapsed, diffuse large B-cell lymphoma (DLBCL). Inotuzumab ozogamicin is currently being tested in phase III clinical trials in acute lymphocytic leukemia (ALL). Particular focus is dedicated to reviewing the pre-clinical and clinical data generated with this compound in NHL and to outline future focus areas for pre-clinical- and clinical research of inotuzumab ozogamicin, and the calicheamicin class of antibody-drug conjugates more generally.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Hematologic Neoplasms/therapy , Immunoconjugates/therapeutic use , Immunotherapy/methods , Sialic Acid Binding Ig-like Lectin 2/immunology , Antibodies, Monoclonal, Humanized/immunology , Cell Proliferation/drug effects , Cell Proliferation/genetics , DNA Breaks, Double-Stranded/drug effects , Drug Screening Assays, Antitumor , Hematologic Neoplasms/genetics , Hematologic Neoplasms/immunology , Humans , Immunoconjugates/immunology , Inotuzumab Ozogamicin , Lymphoma, Non-Hodgkin/genetics , Lymphoma, Non-Hodgkin/immunology , Lymphoma, Non-Hodgkin/therapy
20.
Biochem Pharmacol ; 91(2): 135-43, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-24950467

ABSTRACT

Most oncology compounds entering clinical development have passed stringent preclinical pharmacology evaluation criteria. However, only a small fraction of experimental agents induce meaningful antitumor activities in the clinic. Low predictability of conventional preclinical pharmacology models is frequently cited as a main reason for the unusually high clinical attrition rates of therapeutic compounds in oncology. Therefore, improvement in the predictive values of preclinical efficacy models for clinical outcome holds great promise to reduce the clinical attrition rates of experimental compounds. Recent reports suggest that pharmacology studies conducted with patient derived xenograft (PDX) tumors are more predictive for clinical outcome compared to conventional, cell line derived xenograft (CDX) models, in particular when therapeutic compounds were tested at clinically relevant doses (CRDs). Moreover, the study of the most malignant cell types within tumors, the tumor initiating cells (TICs), relies on the availability of preclinical models that mimic the lineage hierarchy of cells within tumors. PDX models were shown to more closely recapitulate the heterogeneity of patient tumors and maintain the molecular, genetic, and histological complexity of human tumors during early stages of sequential passaging in mice, rendering them ideal tools to study the responses of TICs, tumor- and stromal cells to therapeutic intervention. In this commentary, we review the progress made in the development of PDX models in key areas of oncology research, including target identification and validation, tumor indication search and the development of a biomarker hypothesis that can be tested in the clinic to identify patients that will benefit most from therapeutic intervention.


Subject(s)
Antineoplastic Agents/pharmacology , Heterografts , Neoplasms, Experimental , Animals , Drug Resistance, Neoplasm , Humans , Neoplasms, Experimental/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...