Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters











Database
Language
Publication year range
1.
Br J Haematol ; 185(2): 240-253, 2019 04.
Article in English | MEDLINE | ID: mdl-30767211

ABSTRACT

Chemotherapy plus rituximab has been the mainstay of treatment for follicular lymphoma (FL) for two decades but is associated with immunosuppression and relapse. In phase 2 studies, lenalidomide combined with rituximab (R2 ) has shown clinical synergy in front-line and relapsed/refractory FL. Here, we show that lenalidomide reactivated dysfunctional T and Natural Killer (NK) cells ex vivo from FL patients by enhancing proliferative capacity and T-helper cell type 1 (Th1) cytokine release. In combination with rituximab, lenalidomide improved antibody-dependent cellular cytotoxicity in sensitive and chemo-resistant FL cells, via a cereblon-dependent mechanism. While single-agent lenalidomide and rituximab increased formation of lytic NK cell immunological synapses with primary FL tumour cells, the combination was superior and correlated with enhanced cytotoxicity. Immunophenotyping of FL patient samples from a phase 3 trial revealed that R2 treatment increased circulating T- and NK-cell counts, while R-chemotherapy was associated with reduced cell numbers. Finally, using an in vitro model of myeloid differentiation, we demonstrated that lenalidomide caused a reversible arrest in neutrophil maturation that was distinct from a cytotoxic chemotherapeutic agent, which may help explain the lower rates of neutropenia observed with R2 versus R-chemotherapy. Taken together, we believe these data support a paradigm shift in the treatment of FL - moving from combination immunochemotherapy to chemotherapy-free immunotherapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Lenalidomide/administration & dosage , Lymphoma, Follicular/drug therapy , Rituximab/administration & dosage , Antibody-Dependent Cell Cytotoxicity/drug effects , Antibody-Dependent Cell Cytotoxicity/immunology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation/drug effects , Cyclophosphamide/therapeutic use , Cytokines/biosynthesis , Doxorubicin/therapeutic use , Drug Resistance, Neoplasm/immunology , Humans , Immunological Synapses/drug effects , Immunological Synapses/immunology , Immunotherapy/methods , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Lenalidomide/immunology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Lymphocyte Count , Lymphoma, Follicular/immunology , Neutrophils/drug effects , Prednisone/therapeutic use , Rituximab/immunology , Rituximab/therapeutic use , Tumor Cells, Cultured , Vincristine/therapeutic use
2.
Bioconjug Chem ; 26(5): 919-31, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25915780

ABSTRACT

Sacituzumab govitecan (IMMU-132) is an antibody-drug conjugate (ADC) made from a humanized anti-Trop-2 monoclonal antibody (hRS7) conjugated with the active metabolite of irinotecan, SN-38. In addition to its further characterization, as the clinical utility of IMMU-132 expands to an ever-widening range of Trop-2-expressing solid tumor types, its efficacy in new disease models needs to be explored in a nonclinical setting. Unlike most ADCs that use ultratoxic drugs and stable linkers, IMMU-132 uses a moderately toxic drug with a moderately stable carbonate bond between SN-38 and the linker. Flow cytometry and immunohistochemistry disclosed that Trop-2 is expressed in a wide range of tumor types, including gastric, pancreatic, triple-negative breast (TNBC), colonic, prostate, and lung. While cell-binding experiments reveal no significant differences between IMMU-132 and parental hRS7 antibody, surface plasmon resonance analysis using a Trop-2 CM5 chip shows a significant binding advantage for IMMU-132 over hRS7. The conjugate retained binding to the neonatal receptor, but it lost greater than 60% of the antibody-dependent cell-mediated cytotoxicity activity compared to that of hRS7. Exposure of tumor cells to either free SN-38 or IMMU-132 demonstrated the same signaling pathways, with pJNK1/2 and p21(WAF1/Cip1) upregulation followed by cleavage of caspases 9, 7, and 3, ultimately leading to poly-ADP-ribose polymerase cleavage and double-stranded DNA breaks. Pharmacokinetics of the intact ADC in mice reveals a mean residence time (MRT) of 15.4 h, while the carrier hRS7 antibody cleared at a similar rate as that of the unconjugated antibody (MRT ∼ 300 h). IMMU-132 treatment of mice bearing human gastric cancer xenografts (17.5 mg/kg; twice weekly × 4 weeks) resulted in significant antitumor effects compared to that of mice treated with a nonspecific control. Clinically relevant dosing schemes of IMMU-132 administered either every other week, weekly, or twice weekly in mice bearing human pancreatic or gastric cancer xenografts demonstrate similar, significant antitumor effects in both models. Current Phase I/II clinical trials ( ClinicalTrials.gov , NCT01631552) confirm anticancer activity of IMMU-132 in cancers expressing Trop-2, including gastric and pancreatic cancer patients.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal/chemistry , Antigens, Neoplasm/immunology , Camptothecin/analogs & derivatives , Cell Adhesion Molecules/immunology , Immunoconjugates/pharmacology , Pancreatic Neoplasms/drug therapy , Stomach Neoplasms/drug therapy , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal, Humanized/therapeutic use , Antibody-Dependent Cell Cytotoxicity/drug effects , Antigens, Neoplasm/metabolism , Apoptosis/drug effects , Camptothecin/chemistry , Camptothecin/pharmacokinetics , Camptothecin/pharmacology , Camptothecin/therapeutic use , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Clinical Trials as Topic , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunoconjugates/chemistry , Immunoconjugates/pharmacokinetics , Immunoconjugates/therapeutic use , Irinotecan , Mice , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Signal Transduction/drug effects , Stomach Neoplasms/immunology , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Xenograft Model Antitumor Assays
3.
Mol Pharm ; 12(6): 1836-47, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25402018

ABSTRACT

The antibody-drug conjugate (ADC), IMMU-130, of the moderately cytotoxic topoisomerase I inhibitor, SN-38, and the CEACAM5-targeted humanized antibody (mAb), labetuzumab, was evaluated in model systems of human colon carcinoma and in phase I clinical trials of heavily pretreated patients with metastatic colorectal cancer. The conjugate, designed with a near-homogeneous drug substitution of 7-8 SN-38/mAb and with a linker that released 50% of the drug in ∼20 h, showed significant antitumor effects compared to a nontargeted ADC in human tumor xenografts, which could be augmented in combination with bevacizumab. The advantage of fractionated dosing was demonstrated, with potential implications for the clinical dosing schedule. Biodistribution comparing IMMU-130 with labetuzumab showed that the conjugate cleared somewhat faster from the blood, but this did not affect tumor uptake and retention. The use of an ultrastable linker in the conjugate design abrogated antitumor effects. A tolerability study in rabbits showed a high safety margin, with no-observed-adverse-effect level (NOAEL) corresponding to a cumulative human-equivalent protein dose of 40-60 mg/kg. The preclinical findings appear to be corroborated in two phase I clinical trials, with high tolerability and evidence of antitumor activity, including objective responses. The impact of the ADC design on the utility of IMMU-130, tailored to a poorly internalizing target, is discussed.


Subject(s)
Camptothecin/analogs & derivatives , Immunoconjugates/chemistry , Immunoconjugates/therapeutic use , Animals , Antibodies, Monoclonal , Antineoplastic Agents , Bevacizumab/therapeutic use , Camptothecin/chemistry , Camptothecin/therapeutic use , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Female , Humans , Immunoconjugates/pharmacokinetics , Irinotecan , Mice , Mice, Nude , Rabbits
4.
BMC Cancer ; 13: 170, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23548153

ABSTRACT

BACKGROUND: Advanced or metastatic renal cell carcinoma (RCC) has a poor prognosis, because it is relatively resistant to conventional chemotherapy or radiotherapy. Treatments with human interferon-α2b alone or in combination with mammalian target of rapamycin (mTOR) inhibitors have led to only a modest improvement in clinical outcome. One observation made with mTOR inhibitors is that carcinomas can overcome these inhibitory effects by activating the insulin-like growth factor-I (IGF-I) signaling pathway. Clinically, there is an association of IGF-I receptor (IGF-IR) expression in RCC and poor long-term patient survival. We have developed a humanized anti-IGF-IR monoclonal antibody, hR1, which binds to RCC, resulting in effective down-regulation of IGF-IR and moderate inhibition of cell proliferation in vitro. In this work, we evaluate the anti-tumor activity of two novel IGF-1R-targeting agents against renal cell carcinoma given alone or in combination with an mTOR inhibitor. METHODS: hR1 was linked by the DOCK-AND-LOCK™ (DNL™) method to four Fabs of hR1, generating Hex-hR1, or to four molecules of interferon-α2b, generating 1R-2b. Eight human RCC cell lines were screened for IGF-1R expression and sensitivity to treatment with hR1 in vitro. Synergy with an mTOR inhibitor, temsirolimus, was tested in a cell line (ACHN) with low sensitivity to hR1. RESULTS: Hex-hR1 induced the down-regulation of IGF-IR at 10-fold lower concentrations compared to the parental hR1. Sensitivity to growth inhibition mediated by hR1 and Hex-hR1 treatments correlated with IGF-1R expression (higher expression was more sensitive). The potency of 1R-2b to inhibit the in vitro growth of RCC was also demonstrated in two human cell lines, ACHN and 786-O, with EC50-values of 63 and 48 pM, respectively. When combined with temsirolimus, a synergistic growth-inhibition with hR1, Hex-hR1, and 1R-2b was observed in ACHN cells at concentrations as low as 10 nM for hR1, 1 nM for Hex-hR1, and 2.6 nM for 1R-2b. CONCLUSIONS: Both Hex-hR1 and 1R-2b proved to be more potent than parental hR1 in inhibiting growth of RCC in vitro. Synergy was achieved when each of the three hR1-based agents was combined with temsirolimus, suggesting a new approach for treating RCC.


Subject(s)
Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/metabolism , Protein Kinase Inhibitors/pharmacology , Receptor, IGF Type 1/antagonists & inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Carcinoma, Renal Cell/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation , Drug Resistance, Neoplasm , Drug Synergism , Humans , Kidney Neoplasms/genetics , Protein Kinase Inhibitors/chemistry , Receptor, IGF Type 1/metabolism , TOR Serine-Threonine Kinases/metabolism
5.
PLoS One ; 7(8): e44235, 2012.
Article in English | MEDLINE | ID: mdl-22952934

ABSTRACT

A major mechanism of monoclonal antibodies that selectively target the insulin-like growth factor type 1 receptor (IGF-1R) to inhibit tumor growth is by downregulating the receptor, regardless whether they are capable (antagonistic) or incapable (agonistic) of blocking the binding of cognate ligands. We have developed and characterized a novel agonistic anti-IGF-1R humanized antibody, hR1, and used the Dock-and-Lock (DNL) method to construct Hex-hR1, the first multivalent antibody comprising 6 functional Fabs of hR1, with the aim of enhancing potency of hR1. Based on cross-blocking experiments, hR1 recognizes a region of cysteine-rich domain on the α-subunit, different from the epitopes mapped for existing anti-IGF-1R antibodies, yet hR1 is similar to other anti-IGF-1R antibodies in downregulating IGF-1R and inhibiting proliferation, colony formation, or invasion of selected cancer cell lines in vitro, as well as suppressing growth of the RH-30 rhabdomyosarcoma xenograft in nude mice when combined with the mTOR inhibitor, rapamycin. Hex-hR1 and hR1 are generally comparable in their bioactivities under the in-intro and in-vivo conditions investigated. Nevertheless, in selective experiments involving a direct comparison of potency, Hex-hR1 demonstrated a stronger effect on inhibiting cell proliferation stimulated by IGF-1 and could effectively downregulate IGF-1R at a concentration as low as 20 pM.


Subject(s)
Immunoglobulin G/immunology , Neoplasms/immunology , Receptor, IGF Type 1/immunology , Animals , Antibodies, Blocking/immunology , Antineoplastic Agents/pharmacology , Cadherins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Chromatography, Gel , Chromatography, High Pressure Liquid , Culture Media, Serum-Free , Down-Regulation/drug effects , Epitopes/immunology , Flow Cytometry , Humans , Light , Mice , Molecular Weight , Neoplasm Invasiveness , Neoplasms/pathology , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Scattering, Radiation , Spectrometry, Mass, Electrospray Ionization , Tumor Stem Cell Assay , Vimentin/metabolism
6.
Clin Cancer Res ; 17(10): 3157-69, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21372224

ABSTRACT

PURPOSE: Evaluate the efficacy of an SN-38-anti-Trop-2 antibody-drug conjugate (ADC) against several human solid tumor types, and to assess its tolerability in mice and monkeys, the latter with tissue cross-reactivity to hRS7 similar to humans. EXPERIMENTAL DESIGN: Two SN-38 derivatives, CL2-SN-38 and CL2A-SN-38, were conjugated to the anti-Trop-2-humanized antibody, hRS7. The immunoconjugates were characterized in vitro for stability, binding, and cytotoxicity. Efficacy was tested in five different human solid tumor-xenograft models that expressed Trop-2 antigen. Toxicity was assessed in mice and in Cynomolgus monkeys. RESULTS: The hRS7 conjugates of the two SN-38 derivatives were equivalent in drug substitution (∼ 6), cell binding (K(d) ∼ 1.2 nmol/L), cytotoxicity (IC(50) ∼ 2.2 nmol/L), and serum stability in vitro (t/(½) ∼ 20 hours). Exposure of cells to the ADC demonstrated signaling pathways leading to PARP cleavage, but differences versus free SN-38 in p53 and p21 upregulation were noted. Significant antitumor effects were produced by hRS7-SN-38 at nontoxic doses in mice bearing Calu-3 (P ≤ 0.05), Capan-1 (P < 0.018), BxPC-3 (P < 0.005), and COLO 205 tumors (P < 0.033) when compared to nontargeting control ADCs. Mice tolerated a dose of 2 × 12 mg/kg (SN-38 equivalents) with only short-lived elevations in ALT and AST liver enzyme levels. Cynomolgus monkeys infused with 2 × 0.96 mg/kg exhibited only transient decreases in blood counts, although, importantly, the values did not fall below normal ranges. CONCLUSIONS: The anti-Trop-2 hRS7-CL2A-SN-38 ADC provides significant and specific antitumor effects against a range of human solid tumor types. It is well tolerated in monkeys, with tissue Trop-2 expression similar to humans, at clinically relevant doses, and warrants clinical investigation.


Subject(s)
Antigens, Neoplasm/immunology , Camptothecin/analogs & derivatives , Cell Adhesion Molecules/immunology , Immunoglobulin G/therapeutic use , Neoplasms, Glandular and Epithelial/drug therapy , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Camptothecin/administration & dosage , Camptothecin/chemistry , Camptothecin/therapeutic use , Cell Adhesion Molecules/antagonists & inhibitors , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Immunoconjugates/chemistry , Immunoconjugates/therapeutic use , Immunoglobulin G/chemistry , Irinotecan , Macaca fascicularis , Male , Mice , Mice, Nude , Models, Biological , Monkey Diseases/drug therapy , Monkey Diseases/pathology , Treatment Outcome , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL