621 A Tregs depleting, CD25-targeted antibody-drug conjugate synergizes with tumor-targeted radiotherapy and systemic interleukin-2 in pre-clinical models of solid cancers

2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A657-A657
Author(s):  
Patrick van Berkel ◽  
Francesca Zammarchi

BackgroundRegulatory T cells (Tregs) contribute to an immunosuppressive tumor microenvironment. High tumor infiltration by Tregs and a low ratio of Teffector cells/Tregs is often associated with poor prognosis in solid tumors.1 Tregs represent a major obstacle to cancer immunotherapies, including checkpoint inhibitors and interleukin-2 (IL-2) and are associated with tumors resistance to radiotherapy.2 CD25-ADC (a.k.a. sur301) is an antibody-drug conjugate (ADC) composed of rat monoclonal antibody PC61, directed against mouse CD25, conjugated to tesirine, a pyrrolobenzodiazepine (PBD) dimer-based protease-cleavable linker.3 Previously we showed that single low doses of CD25-ADC resulted in potent and durable antitumor activity in established syngeneic solid tumor models and the combination of a suboptimal dose was synergistic with PD-1 blockade. Tumor eradication by CD25-ADC was CD8+ T cell-dependent and it induced protective immunity. Importantly, while CD25-ADC mediated a significant and sustained intratumoral Tregs depletion, accompanied by a concomitant increase in the number of activated and proliferating tumor-infiltrating CD8+ Teffs cells, systemic Tregs depletion was transient, alleviating concerns of potential autoimmune side effects.4MethodsHere we evaluated the anti-tumor activity of CD25-ADC combined with tumor-targeted radiotherapy (RT) or systemic IL-2 in syngeneic solid tumor models.ResultsTo investigate the combination with radiotherapy, single low doses of CD25-ADC (0.25 or 0.5 mg/kg) were administered intravenously either alone or in combination with image-guided focal radiation in the CT26 syngeneic model. Both doses of CD25-ADC alone induced significant anti-tumor activity compared to the vehicle control. Combination of CD25-ADC, at 0.25 or 0.5 mg/kg, with focal radiotherapy resulted in synergistic anti-tumor activity with 60% and 80% tumor-free survivors (TFS), respectively, at the end of the study. Moreover, re-challenged TFS did not develop new tumors, demonstrating development of tumor-specific protective immunity.The combination of CD25-ADC and systemic IL-2 was investigated in the MC38 syngeneic model. CD25-ADC was administered intravenously either alone (0.25 or 0.5 mg/kg, single dose) or in combination with IL-2 (0.1 or 0.8 mg/kg). Single doses of CD25-ADC elicited significant anti-tumor activity compared to the vehicle control. Combination of CD25-ADC with IL-2 resulted in enhanced anti-tumor activity in both combination groups compared to the respective single agents and the combination was synergistic at the highest IL-2 dose.ConclusionsTogether, these new preclinical data show novel promising combination regimens for CD25-ADC and other commonly used anti-cancer treatments and they provide rationale for the investigation of camidanlumab tesirine (ADCT-301), a PBD-based ADC targeting human CD25, in similar clinical combinations settings.ReferencesShang, B., et al., Prognostic value of tumor-infiltrating FoxP3+ regulatory T cells in cancers: a systematic review and meta-analysis. Sci Rep 2015;5: p. 15179.Liu S, et al., Effects of radiation on T regulatory cells in normal states and cancer: mechanisms and clinical implications. Am J Cancer Res 2015;5(11):p. 3276–85.Zammarchi, F., et al., A CD25-targeted pyrrolobenzodiazepine dimer-based antibody-drug conjugate shows potent anti-tumor activity in pre-clinical models of solid tumors either alone or in combination with a PD-1 inhibitor. Journal for ImmunoTherapy of Cancer 2018. 6(Supplement 1).Zammarchi, F., et al., A CD25-targeted antibody-drug conjugate depletes regulatory T cells and eliminates established syngeneic tumors via antitumor immunity. Journal for ImmunoTherapy of Cancer 2020. In press.

2020 ◽  
Vol 8 (2) ◽  
pp. e000860 ◽  
Author(s):  
Francesca Zammarchi ◽  
Karin Havenith ◽  
Francois Bertelli ◽  
Balakumar Vijayakrishnan ◽  
Simon Chivers ◽  
...  

BackgroundRegulatory T cells (Tregs) contribute to an immunosuppressive tumor microenvironment. They play an important role in the establishment and progression of tumors with high Tregs infiltration and present a major obstacle to tumor eradication by immunotherapies. Numerous strategies have been attempted to deplete or block Tregs, although their success has been limited.MethodsA CD25-targeted, pyrrolobenzodiazepine (PBD) dimer-based antibody–drug conjugate (ADC) was investigated for its ability to deplete Tregs and induce antitumor immunity. Antitumor activity of CD25-ADC either alone or in combination with an anti-programmed cell death protein 1 (PD-1) antibody was evaluated in CD25-negative syngeneic models that exhibit tumor infiltration of CD25-expressing Tregs, and its pharmacodynamics and pharmacokinetics were assessed.ResultsSingle low doses of CD25-ADC resulted in potent and durable antitumor activity in established syngeneic solid tumor models and the combination of a suboptimal dose was synergistic with PD-1 blockade. Tumor eradication by the CD25-targeted ADC was CD8+ T cell-dependent and CD25-ADC induced protective immunity. Importantly, while CD25-ADC mediated a significant and sustained intratumoral Tregs depletion, accompanied by a concomitant increase in the number of activated and proliferating tumor-infiltrating CD8+ T effector cells, systemic Tregs depletion was transient, alleviating concerns of potential autoimmune side effects.ConclusionsThis study shows that a PBD dimer-based, CD25-targeted ADC is able to deplete Tregs and eradicate established tumors via antitumor immunity. This represents a novel approach to efficiently deplete Tregs via a very potent DNA damaging toxin known to induce immunogenic cell death. Moreover, this study provides proof of concept for a completely new application of ADCs as immunotherapeutic agents, as the main mode of action relies on the ADC directly targeting immune cells, rather than tumor cells. These strong preclinical data warrant the clinical evaluation of camidanlumab tesirine (ADCT-301), a PBD-based ADC targeting human CD25, either alone or in combination with checkpoint inhibitors in solid tumors with known Tregs infiltration. A phase I trial (NCT03621982) of camidanlumab tesirine in patients with selected advanced solid tumors is ongoing.


2021 ◽  
pp. molcanther.0708.2020
Author(s):  
Qing Sheng ◽  
Joseph Anthony D'Alessio ◽  
Daniel L. Menezes ◽  
Christopher Karim ◽  
Yan Tang ◽  
...  

2019 ◽  
Vol 449 ◽  
pp. 87-98 ◽  
Author(s):  
Katsuya Nagaoka ◽  
Xuewei Bai ◽  
Kosuke Ogawa ◽  
Xiaoqun Dong ◽  
Songhua Zhang ◽  
...  

Haematologica ◽  
2020 ◽  
Vol 105 (11) ◽  
pp. 2584-2591 ◽  
Author(s):  
Eugenio Gaudio ◽  
Chiara Tarantelli ◽  
Filippo Spriano ◽  
Francesca Guidetti ◽  
Giulio Sartori ◽  
...  

Antibody drug conjugates represent an important class of anti-cancer drugs in both solid tumors and hematological cancers. Here, we report preclinical data on the anti-tumor activity of the first-in-class antibody drug conjugate MEN1309/OBT076 targeting CD205. The study included preclinical in vitro activity screening on a large panel of cell lines, both as single agent and in combination and validation experiments on in vivo models. CD205 was first shown frequently expressed in lymphomas, leukemias and multiple myeloma by immunohistochemistry on tissue microarrays. Anti-tumor activity of MEN1309/OBT076 as single agent was then shown across 42 B-cell lymphoma cell lines with a median IC50 of 200 pM and induction of apoptosis in 25/42 (59.5%) of the cases. The activity appeared highly correlated with its target expression. After in vivo validation as the single agent, the antibody drug conjugate synergized with the BCL2 inhibitor venetoclax, and the anti-CD20 monoclonal antibody rituximab. The first-in-class antibody drug targeting CD205, MEN1309/OBT076, demonstrated strong pre-clinical anti-tumor activity in lymphoma, warranting further investigations as a single agent and in combination.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 5623-5623
Author(s):  
Andrew Hau ◽  
Tong Zhu ◽  
Rengang Wang ◽  
Megan Lau ◽  
Lingna Li ◽  
...  

BCMA (B-cell maturation antigen) is an integral membrane protein that belongs to the TNF receptor family with expression restricted to B cell lineage cells. The RNA is near universally detected in multiple myeloma (MM) cells and the protein is expressed on the surface of malignant plasma cells from patients with MM. In contrast, BCMA expression in normal tissues is very limited, making BCMA a promising target for antibody-drug conjugate (ADC) therapy. We have developed a BCMA-targeting ADC, employing a fully human anti-BCMA monoclonal antibody (mAb) identified from Sorrento's G-MAB antibody library, which was conjugated using proprietary Concortis linker-Duo 5.2 toxin technology resulting in BCMA-077 ADC. The mAb has a unique binding profile for BCMA and demonstrated strong preferential binding for BCMA-overexpressing cells but showed much less binding to lower BCMA-expressing cells. This property allows for more selective binding of the ADC on high BCMA-expressing cells, which are usually tumor cells while sparing low BCMA-expressing normal cells. In addition, we modified the Duo 5.2 payload decreasing the potency of the unconjugated toxin while retaining activity when conjugated to the mAb. The resulting ADC, BCMA-024, was compared to BCMA-077 using in vitro assays, including binding, internalization and cytotoxicity against tumor cell lines. The two ADCs exhibited strong activity and no difference in cytotoxic potency evident. The toxicity of the payload derivative was evaluated in a rodent model and it was found to be well tolerated not showing toxicity at a dose 10 times higher than the lethal dose of the parental toxin. Both ADCs carrying either the parental Duo 5.2 toxin or the derivative toxin payload were evaluated in vivo for anti-tumor activity in three different multiple myeloma xenograft models using different dose regimens. The data showed that both ADCs demonstrated potent BCMA-dependent in vivo anti-tumor activity in all xenograft BCMA-positive tumor models. The PK of the parental anti-BCMA mAb was investigated in non-human primates (NHP) and the parameters indicated a T1/2 of about 10 days. The GLP toxicity studies are ongoing. Our BCMA-ADCs have shown favorable anti-tumor activities combined with good safety profiles resulting in an expanded therapeutic window. The data make BCMA-077 and BCMA-024 promising candidates for continued preclinical development. Based on the totality of our preclinical data, we anticipate selecting a BCMA ADC clinical candidate for the treatment of multiple myeloma. Disclosures Hau: Concortis Biotherapeutics: Employment, Equity Ownership. Zhu:Levena Biopharma: Employment, Equity Ownership, Patents & Royalties. Wang:Concortis Biotherapeutics: Employment, Equity Ownership. Lau:Levena Biopharma: Employment, Equity Ownership. Li:Concortis Biotherapeutics: Employment, Equity Ownership. Li:Levena Biopharma: Employment, Equity Ownership. Sun:Levena Biopharma: Employment, Equity Ownership. Kovacs:Levena Biopharma: Employment, Equity Ownership. Khasanov:Levena Biopharma: Employment, Equity Ownership. Deng:Levena Biopharma: Employment, Equity Ownership. Yan:Levena Biopharma: Employment, Equity Ownership. Knight:Sorrento Therapeutics, Inc.: Employment, Equity Ownership. Kaufmann:Sorrento Therapeutics, Inc.: Employment, Equity Ownership, Patents & Royalties. Ji:Sorrento Therapeutics Inc: Employment, Equity Ownership, Patents & Royalties; Celularity, Inc.: Equity Ownership, Membership on an entity's Board of Directors or advisory committees. Li:Levena Biopharma: Employment, Equity Ownership, Patents & Royalties; Sorrento Therapeutics, Inc.: Employment, Equity Ownership, Patents & Royalties. Zhang:Concortis Biotherapeutics: Employment, Equity Ownership, Patents & Royalties.


Sign in / Sign up

Export Citation Format

Share Document