Abstract P6-15-15: MM-111 - A Novel Bispecific Antibody Targeting HER-2/HER-3 Z Heterodimer: Safety and Tolerability in a First-In Human Phase I/II Study in Patients with Refractory HER2-Positive (HER-2+) Cancers

Author(s):  
M Beeram ◽  
CS Denlinger ◽  
AW Tolcher ◽  
LJ Goldstein ◽  
A Patnaik ◽  
...  
2018 ◽  
Vol 29 ◽  
pp. viii408-viii409 ◽  
Author(s):  
M. Wermke ◽  
J. Alt ◽  
J. Kauh ◽  
J. Back ◽  
Y. Salhi ◽  
...  

2018 ◽  
Vol 82 (3) ◽  
pp. 407-418 ◽  
Author(s):  
Amita Patnaik ◽  
Michael Gordon ◽  
Frank Tsai ◽  
Kyri Papadopoulous ◽  
Drew Rasco ◽  
...  

2001 ◽  
Vol 24 (1) ◽  
pp. 79-87 ◽  
Author(s):  
Thomas Schwaab ◽  
Lionel D. Lewis ◽  
Bernard F. Cole ◽  
Yashwant Deo ◽  
Michael W. Fanger ◽  
...  

2012 ◽  
Vol 30 (15_suppl) ◽  
pp. TPS663-TPS663 ◽  
Author(s):  
Pamela N. Munster ◽  
Kathy Miller ◽  
Ian E. Krop ◽  
Navreet Dhindsa ◽  
Joe Reynolds ◽  
...  

TPS663 Background: Anthracyclines have been an effective backbone of breast cancer therapies for decades. However, cardiotoxicity issues associated with free anthracyclines have limited their effective use in the clinic and led to the exploration of anthracycline-free regimens, particularly with HER2-positive cancers that require treatment with another cardiotoxic agent, trastuzumab. While liposomal doxorubicin formulations have succeeded in reducing cardiotoxicity, they have failed to demonstrate clear-cut efficacy advantages and can involve other toxicities. To address the safety and efficacy limitations of currently available anthracyclines, we have designed a new liposomal formulation, MM-302, that targets doxorubicin to HER2-overexpressing tumor cells. Antibody fragments that bind to HER2 without blocking HER2-mediated signaling are coupled to the outer surface of pegylated liposomal doxorubicin. MM-302 specifically binds and enters tumor cells overexpressing HER2 with minimal uptake into normal cells such as cardiomyocytes which express low levels of HER2. This first-in-human phase I study evaluates the safety of MM-302 in patients and provides preliminary efficacy data in HER-2+ advanced breast cancer (ABC). Methods: Patients aged > 18 years with histologically confirmed HER-2+ advanced breast cancer that have progressed or recurred on standard therapy or for which no standard therapy exists who have adequate performance status, bone marrow reserve, and organ function, are eligible for the study. Following a standard 3 + 3 dose escalation design, the maximum tolerated dose (MTD) or maximum feasible dose (MFD) is determined and up to 25 additional patients with HER-2+ ABC will be enrolled for a planned total of 40-49 patients. The primary endpoint is determination of the MTD/MFD. Secondary endpoints include determination of dose-limiting toxicity, adverse event(s), and pharmacokinetic and immunogenicity profiles of MM-302, as well as overall response and clinical benefit rates of MM-302. MM-302 is administered intravenously weekly in 4-week cycles. At the time of this submission, 8 patients have been enrolled in the dose escalation portion.


Author(s):  
Emiliano Calvo ◽  
Maria Alsina ◽  
Jan H.M. Schellens ◽  
Alwin DR Huitema ◽  
Josep Tabernero ◽  
...  

2018 ◽  
Vol 82 (3) ◽  
pp. 419-419
Author(s):  
Amita Patnaik ◽  
Michael Gordon ◽  
Frank Tsai ◽  
Kyriakos P. Papadopoulos ◽  
Drew Rasco ◽  
...  

2006 ◽  
Vol 24 (18_suppl) ◽  
pp. 2572-2572 ◽  
Author(s):  
L. G. Salazar ◽  
J. L. Murray ◽  
M. L. Disis ◽  
M. Cheever

2572 Background: Standard methods for immunization with peptides to elicit cytotoxic T lymphocytes (CTL) have not been well defined. Injection of soluble antigens in most standard vaccine formulations induce helper T cell and antibody responses, but generally fail to elicit CTL. Preclinical studies have shown that peptides incorporated into Poly-lactide-co-glycolide (PLG) microspheres are more effective in eliciting CTL than soluble peptides. Additionally, studies in HLA-A2 transgenic mice have shown GM-CSF and monophosphoryl lipid A (MPL-AF) to be effective adjuvants for induction of CTL with PLG peptide. A Phase I vaccine study was conducted to evaluate the safety and immunogenicity of HER2 peptide p369–377 incorporated in PLG microspheres in combination with GM-CSF or MPL-AF as adjuvant in patients with advanced stage HER2 overexpressing cancers. Methods: Twenty-four HLA-A2+ patients with stage III/IV breast, ovarian, or non-small cell lung cancer were enrolled sequentially into 4 treatment arms (6 patients/arm) and received escalating doses of the vaccine (Arms 1 and 2 received 0.5 mg of HER2 peptide; Arms 3 and 4 received 1.5 mg HER2 peptide) with different adjuvants (Arms 1, 2 and 3 received GM-CSF; Arm 4 received MPL-AF) and by different administration routes (Arm 1 received intradermal injection; Arms 2, 3, and 4 received subcutaneous injection). Vaccines were administered every 28 days for a total of 6 immunizations. Toxicity assessments were conducted at baseline, prior to each vaccine and at follow-up. Immunologic responses were evaluated with IFN-γ ELIspot assay. Results: Fourteen subjects completed all 6 vaccinations. 7 patients withdrew from study because of disease progression. No serious or grade 4 toxicity related to vaccine occurred. Eighteen of 24 subjects were evaluable for immunologic responses. 11/18 subjects (61%) developed a p369–377-specific CTL response. Immune responses did not appear to be related to treatment arm. Conclusions: The HER2 p369–377/PLG vaccine plus adjuvant is safe and elicits HER2-specific T cells in patients with HER2 positive cancers. No significant financial relationships to disclose.


Sign in / Sign up

Export Citation Format

Share Document