scholarly journals Antibody-dependent cellular cytotoxicity of the optimized anti-CD20 monoclonal antibody ublituximab on chronic lymphocytic leukemia cells with the 17p deletion

Leukemia ◽  
2013 ◽  
Vol 28 (1) ◽  
pp. 230-233 ◽  
Author(s):  
M Le Garff-Tavernier ◽  
L Herbi ◽  
C de Romeuf ◽  
F Nguyen-Khac ◽  
F Davi ◽  
...  
Blood ◽  
2008 ◽  
Vol 112 (13) ◽  
pp. 5180-5189 ◽  
Author(s):  
Rosa Lapalombella ◽  
Bo Yu ◽  
Georgia Triantafillou ◽  
Qing Liu ◽  
Jonathan P. Butchar ◽  
...  

Abstract Lenalidomide, an immunomodulatory agent that enhances antibody-dependent cellular cytotoxicity (ADCC), is currently being investigated as a therapy for chronic lymphocytic leukemia (CLL). The anti-CD20 antibody rituximab is active in CLL and represents a rational agent to combine with lenalidomide. We therefore examined whether lenalidomide combined with rituximab enhances direct apoptosis and ADCC in CLL cells. In contrast to previous reports using CD20-positive lymphoma cell lines, lenalidomide down-regulated CD20 surface antigen expression in CLL patient cells via enhanced internalization, without influencing transcription. The CD20 surface antigen internalization enhanced delivery of an oligonucleotide incorporated into anti-CD20 immunoliposomes. In addition, CD20 surface antigen down-modulation by lenalidomide in CLL was accompanied by diminished rituximab-mediated apoptosis and ADCC. These observations suggest a need for alternative sequencing strategies to avoid antagonism between lenalidomide and rituximab therapy in CLL. In addition, they suggest that lenalidomide therapy might be useful to enhance targeted delivery of RNAi-based therapies using CD20 immunoliposomes in B-cell malignancies.


1994 ◽  
Vol 39 (2) ◽  
pp. 137-146 ◽  
Author(s):  
P. Tassone ◽  
P. Bonelli ◽  
F. Tuccillo ◽  
H.M. Bond ◽  
M.C. Turco ◽  
...  

2019 ◽  
Vol 8 (2) ◽  
pp. IJH14
Author(s):  
Stefano Molica

There were a number of important updates and advances presented at the 2018 Annual American Society of Hematology meeting. With respect to the treatment of chronic lymphocytic leukemia, the American Society of Hematology 2018 was notable for an improved understanding of ibrutinib-based therapies. In fact, three prospective Phase III trials presented at the meeting indicate, in turn, that ibrutinib alone, ibrutinib plus rituximab, or ibrutinib plus obinutuzumab, should be the new standard of care for chronic lymphocytic leukemia. However, additional clinical trials comparing chemo-immunotherapy with ibrutinib alone or in association with an anti-CD20 monoclonal antibody remain a reasonable avenue to complete results of these large studies.


2020 ◽  
Vol 13 (4) ◽  
pp. 221-229
Author(s):  
Caitlin M. McNulty ◽  
Emasenyie A. Isikwei ◽  
Pragya Shrestha ◽  
Melissa R. Snyder ◽  
Brian F. Kabat ◽  
...  

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 984-984
Author(s):  
Bing CUi ◽  
George F. Widhopf ◽  
Jian Yu ◽  
Daniel Martinez ◽  
Esther Avery ◽  
...  

Abstract Abstract 984 ROR1 is an orphan receptor tyrosine kinase that is expressed on leukemia cells of patients with chronic lymphocytic leukemia (CLL), but not on most adult tissues of healthy adults, including CD5+ B cells. To generate anti-ROR1 antibodies, we immunized mice using different strategies employing vaccines comprised of recombinant ROR1 protein, polynucleotide-ROR1 vaccines and CD154 genetic adjuvants, or replication-defective adenovirus vectors encoding ROR1 and CD154. We extirpated the spleens of animals that developed high-titer serum anti-ROR1 antibodies and used these to generate monoclonal-antibody-(mAb)-producing hybridomas or antibody phage-display libraries that subsequently were screened for ROR1-binding. Over 70 unique mAbs were generated that each bound the extra-cellular domain of native ROR1. Most mAbs recognized an epitope(s) within the ROR1 Ig-like domain, which appears to represent the immune dominant epitope. Other mAb recognized epitopes within the conserved ROR1 Kringle domain. One mAb (UC D10-001) had distinctive binding to an intradomain epitope of human ROR1 (hROR1). UC D10-001 was the only mAb we found directly cytotoxic for hROR1-expressing leukemia cells cultured in media without complement for 6 hours. We found that UC D10-001 could induce significant reductions in basal levels of phosphorylated AKT in hROR1-expressing leukemia cells. Moreover, UC D10-001 significantly decreased the basal levels of phosphorylated AKT in freshly isolated human CLL cells (N=4) to levels comparable to that observed in co-cultures containing 10 mM LY294002, a broad-spectrum inhibitor of PI3K. We examined whether this mAb had cytotoxic activity for leukemia cell in vivo. For this we examined whether we could inhibit the adoptive transfer of human-ROR1-expressing leukemia cells to young, syngeneic recipient mice made transgenic for human ROR1 under control of a B-cell specific promoter. Cohorts of 5 animals per group were each given intravenous injections of antibody at a dose of at 10 mg/kg. Each cohort was treated with UC D10-001, control IgG, or 4A5, an anti-ROR1 mAb specific for a non-cross-reactive epitope located in the Ig-like domain of ROR1. Each animal received an intravenous injection of 5 × 105 ROR1-expressing leukemia cells and then was assessed weekly for circulating leukemia cells by flow cytometry. UC D10-001, but not control IgG or 4A5, significantly inhibited engraftment of the ROR1+ leukemia. Four weeks after adoptive transfer, animals treated with UC D10-001 had a 10-fold lower median number of leukemia B cells in the blood than animals treated with control IgG or 4A5. We also tested UC D10-001 for its capacity to induce clearance of human ROR1+ CLL cells engrafted into the peritoneal cavity of Rag-2−/−/γc−/− immune deficient mice. Each of these mice received intraperitoneal injections of equal numbers of human ROR1+ CLL cells prior to receiving D10-001, control IgG, or 4A5, each at 10 mg/kg. These animals were sacrificed seven days later and the human leukemia cells were harvested via peritoneal lavage. In mice treated with UC D10-001 we harvested an average of only 6 × 104 ± 3 × 104 CLL cells. This number of cells was significantly less than the average number of CLL cells harvested from control IgG or 4A5-treated mice (8 × 105 ± 4 × 105 or 7 × 105 ± 2 × 105, respectively, p <0.01). These studies indicate that the anti-ROR1 mAb UC D10-001 can be directly cytotoxic for ROR1-expressing leukemia cells in vitro and in vivo, a property that apparently is unique to this mAb among other anti-ROR1 mAbs. Because of the restricted expression of ROR1 on leukemia cells and the distinctive properties of this mAb, we propose that UC D10-001 might have potential utility in the treatment of patients with CLL. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3886-3886
Author(s):  
Eva Hellqvist ◽  
Christina C.N. Wu ◽  
George F. Widhopf ◽  
Alice Shih ◽  
Rommel Tawatao ◽  
...  

Abstract Abstract 3886 ROR1 is a receptor-tyrosine kinase like protein expressed on the surface of chronic lymphocytic leukemia (CLL) B cells, but not on normal mature B cells, suggesting that it may be a promising therapeutic target. We have generated a chimeric monoclonal antibody (mAb), UC99961, which binds to an intradomain epitope of human ROR1 (hROR1). UC99961 binds the same epitope as the murine anti-hROR1 mAb, UC D10–001, which has direct cytotoxic effects on hROR1 positive CLL cells. In this study we investigated the in-vivo anti-leukemic activity and tolerability of UC99961 on ROR1+ primary patient CLL cells and human cord-blood-derived B cells and T cells, respectively. For these studies, immunodeficient RAG2−/−γc−/− neonatal mice were reconstituted with a human immune system by intrahepatic xenotransplantation of 1×105 CD34+ human cord blood progenitor cells. Eight to ten weeks post transplantation, cord blood engraftment was verified by peripheral blood screening, at which point the mice received an intraperitoneal transplantation of 2×107 primary patient ROR1+ CLL cells. Twenty-four hours after CLL transplantation, five animals per group were each treated with a single intraperitoneal injection (10mg/kg) of UC99961, UC D10–001, or control IgG. Seven days following mAb treatment, the animals were sacrificed and marrow, spleen, thymus, and peritoneal lavage samples were collected and analyzed by flow cytometry for CLL cells, as well as normal cord-blood-derived B cells and T cells. To confirm mAb administration according to the study design, serial residual ROR1 plasma antibody levels were determined by ELISA. Results from three consecutive experiments using leukemia cells from two different patients showed that the vast majority of CLL B cells remained in the peritoneal cavity of the animals and did not migrate to other hematopoietic organs. Both anti-hROR1 mAbs UC99961 and UC D10–001 significantly reduced the average number of harvested CLL cells in the peritoneal lavage compared to control IgG (99% and 71% reduction respectively), while cord-blood-derived T cells (CD45+3+) in thymus remained unaffected by the mAb treatment. For the majority of cord-blood-derived B cells in marrow and spleen, no significant reduction could be observed after UC99961 or UC D10–001 mAb treatment. A small CD19+ROR1+CD34− cord-blood-derived B cell population was identified in marrow and spleen that was reduced after UC99961 and UC D10–001 mAb treatment. This study demonstrates that the anti-human ROR1 specific mAbs have in vivo anti-leukemic activity with minimal impact on human cord-blood-derived B cells and T cells. From these results, UC99961 appears to be an excellent candidate antibody for future clinical studies for patients with CLL. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2873-2873
Author(s):  
George F. Widhopf ◽  
Bing Cui ◽  
Christina C.N. Wu ◽  
Jian Yu ◽  
Fitzgerald Lao ◽  
...  

Abstract ROR1 is an onco-embryonic antigen that is expressed on the neoplastic cells of patients with chronic lymphocytic leukemia (CLL), other B-cell lymphomas, acute leukemias, or many different solid-tumors, but not on non-neoplastic post-partum tissues, except for the uncommon precursor B cells known as hematogones. We generated over 70 hybridomas, each producing a monoclonal-antibody (mAb) specific for the extracellular domain of ROR1 and found only one (D10) that had anti-leukemia activity in a niche-dependent assay, despite having a relatively low ROR1-binding affinity (Kd 40 nM). We generated high-affinity mAbs specific for the epitope recognized by D10 using recombinant phage-display libraries and found one (designated 961) that bound ROR1 with high affinity (Kd 800 pM) and had similar anti-leukemia activity as D10. To mitigate immunogenicity, we identified both light and heavy chain complementary determining regions (CDR) and framework junctions in the 961 mAb. Using conservative CDR and parallel framework substitutions (BioAtla-San Diego) we generated a panel of 21 humanized 961 variants. We selected one (cirmtuzumab or UC-961) that had high specificity and affinity for ROR1 (Kd 4nM). Moreover, UC-961 does not react with normal adult tissues, as assessed by immunohistochemical studies on FDA normal tissue arrays or fresh-frozen adult tissues, but reacted strongly with ROR1+ neoplastic cells (e.g. CLL cells or solid-tumor tissues). We inserted the optimized genetic sequence of the UC-961 mAb into selectable expression cassettes and used these to transfect Chinese hamster ovary cells (CHO-Selexis, Switzerland). Upon successive rounds of single cell cloning, we identified a single clone (Acp7) that stably expressed greater that 2 g/L of UC-961. In pilot studies, we have expanded this clone to over 50 population doublings without change in production quantity or quality of UC-961 during prolonged expansion. Acp7 has been banked, tested, released and used to produce pilot-scale material for process validation, GLP tissue cross and pre-clinical pharmacology/ toxicology testing. We have developed a scalable, 5-step process that generates a purified antibody that results in the removal of >12 logs of test viruses in clearance studies and have transitioned Acp7 into GMP manufacturing (Pacific GMP-San Diego), using a Wave Reactor (GE). From this, we generated approximately 1 g/L of the UC-961 mAb with an overall yield of >75% for the final formulated product. The UC-961 mAb has similar, if not greater, anti-leukemia activity as D10 in our niche-dependent assays. For this we assessed whether the UC-961 anti-human ROR1 mAb could effect clearance of human-ROR1 expressing murine leukemia cells engrafted in immunodeficient recipient mice. Groups of eight RAG2-/-γc-/- mice were each injected intravenously with 0, 3, 10, or 30 mg/kg of UC-961 and then given an intravenous injection of 1x104 CD5+B220lo human ROR1+ murine leukemia cells derived from a ROR1xTCL1 transgenic mouse. Treatment with UC-961 mAb resulted in a 95% clearance of leukemic cells in the spleen at all doses tested, compared to control animals (p <0.01). We also tested UC-961 mAb for its capacity to induce clearance of human ROR1+ CLL cells engrafted into the peritoneal cavity of Rag-2-/-/γc-/- immune deficient mice. For this, mice received a single dose of UC-961 (30, 10, 3, 1, and 0.3 mg/kg) or control vehicle one day after engraftment. Seven days later, the CLL cells were harvested peritoneal lavage, counted, and analyzed by flow cytometry. In a representative experiment, the UC-961 significantly reduced the average number of harvested CLL cells in the peritoneal lavage in a dose dependent manner compared to controls (92% ± 4%, 84% ± 5%, 71% ± 8%, 69% ±14% and 60% ± 10% reduction, respectively, p < 0.001, n = 6 per group), demonstrating the in vivo anti-leukemic activity of the ROR1 targeted mAb. Pharmacokinetic and toxicology studies in Wistar rats have yet to demonstrate dose-limiting toxicity. A one compartment PK description of average data reveals that t1/2 = 11.4 days V= 1.18 mL (47 mL/kg) and CL = 0.072 mL/day (0.12 mL/hr/kg). Collectively, these studies indicate that UC-961 may be suitable for clinical studies in patients with CLL or other ROR1-expressing cancers. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document