Pieris Pharmaceuticals Presents Updated Phase 1 Monotherapy Data for 4-1BB/HER2 Bispecific Cinrebafusp Alfa and Preclinical Data for 4-1BB/PD-L1 Bispecific PRS-344/S095012 at 2021 AACR Annual Meeting

On April 10, 2021 Pieris Pharmaceuticals, Inc. (NASDAQ:PIRS), a clinical-stage biotechnology company advancing novel biotherapeutics through its proprietary Anticalin technology platform for respiratory diseases, cancer, and other indications, reported a clinical data update from the phase 1 monotherapy study of cinrebafusp alfa (PRS-343), a 4-1BB/HER2 bispecific for the treatment of HER2-expressing solid tumors, in an oral presentation at the American Association for Cancer Research (AACR) (Free AACR Whitepaper) Virtual Congress 2021 (Press release, Pieris Pharmaceuticals, APR 10, 2021, View Source [SID1234577822]). The Company also presented preclinical data for PRS-344/S095012, a 4-1BB/PD-L1 bispecific the Company is developing with Servier, at a poster session at the congress.

Schedule your 30 min Free 1stOncology Demo!
Discover why more than 1,500 members use 1stOncology™ to excel in:

Early/Late Stage Pipeline Development - Target Scouting - Clinical Biomarkers - Indication Selection & Expansion - BD&L Contacts - Conference Reports - Combinatorial Drug Settings - Companion Diagnostics - Drug Repositioning - First-in-class Analysis - Competitive Analysis - Deals & Licensing

                  Schedule Your 30 min Free Demo!

Cinrebafusp Alfa (PRS-343):

Presented data demonstrated additional clinical benefit at the highest dose, including an additional, ongoing confirmed durable partial response, three additional patients with stable disease as best response, and overall durable benefit. Based on clinical benefit and pharmacodynamic correlates, cinrebafusp alfa showed a clear dose response and a 4-1BB-driven mechanism of action. Additionally, clinical benefit was observed in patients with "cold" tumors as well as those with HER2-low expressing tumors. Cinrebafusp alfa continues to be well-tolerated. The Company plans to initiate a phase 2 study in gastric cancer this summer that will evaluate both HER2-high and HER2-low patient settings.

As of the cut-off date of February 25, 2021, 8 patients in the monotherapy trial were evaluable for a response at the highest dose cohort (cohort 13b; 18 mg/kg Q2W) out of a total of 42 response-evaluable patients enrolled in the predicted active dose cohorts (cohort 9 and higher; ≥2.5 mg/kg) in the study.

In cohort 13b, one additional patient (cancer of unknown primary) achieved an ongoing confirmed durable partial response, for an updated overall response rate (ORR) of 25% in that cohort as compared to an ORR of 12% across active dose levels.
In cohort 13b, three additional patients experienced stable disease as best response, for an updated disease control rate (DCR) of 63% in that cohort as compared to a DCR of 52% across active dose levels.
Cinrebafusp alfa activates adaptive and innate immunity in the tumor microenvironment, consistent with intended mode of action as evidenced by post-treatment increases in CD8+ T cells, NK cells and cytotoxic activity.
Dose-dependent increases of CD8+ T cells in the tumor and soluble 4-1BB in the blood of patients demonstrate target engagement and a 4-1BB-driven mode of action.
Cinrebafusp alfa shows preliminary evidence of activity among "cold" tumor types as well as "hot" tumor types.
Activity in HER2-low expressing patients supports continued development of cinrebafusp alfa in that population, which the Company will evaluate in its phase 2 gastric cancer study.
Cinrebafusp alfa monotherapy appeared to be well-tolerated up to 18 mg/kg, with no significant specific anti-HER2 or anti-4-1BB safety signal and no dose limiting toxicity identified.
The cinrebafusp alfa data presented at AACR (Free AACR Whitepaper) can be found in an updated corporate presentation at View Source

PRS-344S095012:

The synergistic preclinical data presented for PRS-344/S095012 demonstrate PRS-344/S095012 is superior to the combination of PD-L1- and 4-1BB-targeting molecules. In an anti-PD-L1-resistant mouse model, the drug candidate induces a dose-dependent anti-tumor response and significantly extends survival. In vitro, PRS-344/S095012 enhances effective CD8+ T cell response and proinflammatory cytokine release.

PRS-344/S095012-mediated 4-1BB activation is strictly PD-L1 dependent, reducing the risk of peripheral toxicity. Furthermore, 4-1BB co-stimulation only occurs in combination with simultaneous TCR signaling, restricting its activity to antigen-specific T cells. PRS-344/S095012 also displays mAb-like pharmacokinetics in mice.

These data support further development of PRS-344/S095012, for which the phase 1 study is expected to begin this year.

A copy of the poster is available at this link.

"The matured data from the highest dose cohort of cinrebafusp alfa demonstrate a clear dose-dependent response that supports our recommended phase 2 dose, and the biomarker data generated across all active dose cohorts demonstrate that cinrebafusp alfa activity is 4-1BB-driven and that the drug candidate is active not only in HER2-high expressing tumors, but also HER2-low expressing tumors – a significant opportunity and unmet medical need that we are excited to pursue in our upcoming phase 2 study," said Stephen S. Yoder, President and Chief Executive Officer of Pieris. "Separately, we are pleased with the clear evidence of dose-dependent synergistic anti-tumor effects of PRS-344/S095012, as well as further evidence for its tumor-localized mechanism of action, and we look forward to moving this asset into the clinic this year. By its design, this bispecific has best-in-class potential in the 4-1BB/PD-L1 arena."

About Cinrebafusp Alfa:

Cinrebafusp alfa (PRS-343) is a 4-1BB/HER2 fusion protein comprising a 4-1BB-targeting Anticalin protein and a HER2-targeting antibody. The drug candidate is currently in development for the treatment of HER2-positive solid tumors. Based on encouraging phase 1 study results, which demonstrated clinical benefit as single agent and biomarker data indicative of a 4-1BB-driven mechanism of action, the Company is actively working towards initiating a phase 2 study of cinrebafusp alfa in combination with ramucirumab and paclitaxel for the treatment of HER2-high expressing gastric cancer and in combination with tucatinib in HER2-low expressing gastric cancer.

Synlogic Presents Data from SYNB1891 Phase 1 Trial at American Association for Cancer Research (AACR) Annual Meeting

On April 10, 2021 Synlogic, Inc. (Nasdaq: SYBX), a clinical stage company bringing the transformative potential of synthetic biology to medicine, reported data on SYNB1891 for the treatment of solid tumors and lymphoma during the American Association for Cancer Research (AACR) (Free AACR Whitepaper) annual meeting, April 10-15, 2021 (Press release, Synlogic, APR 10, 2021, View Source [SID1234577821]).

Schedule your 30 min Free 1stOncology Demo!
Discover why more than 1,500 members use 1stOncology™ to excel in:

Early/Late Stage Pipeline Development - Target Scouting - Clinical Biomarkers - Indication Selection & Expansion - BD&L Contacts - Conference Reports - Combinatorial Drug Settings - Companion Diagnostics - Drug Repositioning - First-in-class Analysis - Competitive Analysis - Deals & Licensing

                  Schedule Your 30 min Free Demo!

The presentation, "Intratumoral injection of SYNB1891, a Synthetic Biotic designed to activate the innate immune system, demonstrates target engagement in humans including intratumoral STING activation," was delivered by Dr. Filip Janku, Associate Professor, Department of Investigational Cancer Therapeutics, Division of Cancer Medicine at The University of Texas MD Anderson Cancer Center. The presentation recording will be available throughout the duration of the conference.

SYNB1891 is an investigational drug being evaluated in an ongoing Phase 1 clinical trial for the treatment of solid tumors and lymphoma. SYNB1891 is composed of an engineered Synthetic Biotic strain of E. coli Nissle that produces cyclic di-AMP (CDA), a stimulator of the STING (STimulator of INterferon Genes) pathway. This mechanism can play a critical role in the initiation of an anti-tumor immune response via activation of APCs and presentation of tumor antigens. Findings from the monotherapy cohorts include:

SYNB1891 is safe and well-tolerated as an intratumoral injection in a heterogenous population.
No dose limiting toxicities or SYNB1891-related infections
Dose levels through 1e7 live cells demonstrate target engagement as assessed by dose-dependent increases in serum cytokines, upregulation of ISGs and presence of tumor infiltrating lymphocytes.
Evidence of durable stable disease was seen in 2 patients and was associated with upregulation genes tied to immune activation and increased intratumoral lymphocytes.
These data support continued dose escalation in the monotherapy and combination arms. The combination arm of the study combines escalating dose levels of SYNB1891 with a fixed dose of a PD-L1 checkpoint inhibitor antibody to establish a recommended Phase 2 dose for the combination regimen.

Data from both arms will continue to be reported over the course of 2021, with mature combination therapy data expected by the end of the year.

Learn more about Synlogic’s programs and pipeline by visiting View Source

About SYNB1891
SYNB1891 is an investigational drug for the intra-tumoral treatment of solid tumors and lymphoma, composed of an engineered Synthetic Biotic strain of E. coli Nissle that produces cyclic di-AMP (CDA), a stimulator of the STING (STimulator of INterferon Genes) pathway. This mechanism can play a critical role in the initiation of an anti-tumor immune response via activation of APCs and presentation of tumor antigens. The bacterial chassis of SYNB1891 also stimulates the innate immune system by several other mechanisms, including via Toll-like receptors (TLRs), potentially adding to the magnitude of the overall immune response. While SYNB1891 has been engineered with safety features that are designed to prevent its replication unless supplemented with specific nutrients, the bacteria remain active for several days within the injected tumor to stimulate a local immune response. SYNB1891 is being evaluated in a Phase 1 clinical trial (NCT04167137).

Theratechnologies Presents New In Vivo Pre-Clinical Data At AACR Demonstrating Significant Anti-Tumor Activity Of TH1902 In All Studied Cancer Types

On April 10, 2021 Theratechnologies Inc. (Theratechnologies) (TSX: TH) (NASDAQ: THTX), a biopharmaceutical company focused on the development and commercialization of innovative therapies, reported that new in vivo preclinical data were presented at the 2021 Annual Meeting of the American Association for Cancer Research (AACR) (Free AACR Whitepaper) (Press release, Theratechnologies, APR 10, 2021, View Source [SID1234577820]). These data demonstrated sustained tumor regression, better anti-tumor activity and tolerability with TH1902 compared to docetaxel alone in all cancer types studied, namely melanoma, pancreatic, ovarian, endometrial, colorectal and triple-negative breast cancers. The anti-tumor effect of TH1902 persisted longer post-treatment than with docetaxel alone. TH1902 is the Company’s lead investigational peptide-drug conjugate (PDC) derived from its SORT1+ Technology.

Schedule your 30 min Free 1stOncology Demo!
Discover why more than 1,500 members use 1stOncology™ to excel in:

Early/Late Stage Pipeline Development - Target Scouting - Clinical Biomarkers - Indication Selection & Expansion - BD&L Contacts - Conference Reports - Combinatorial Drug Settings - Companion Diagnostics - Drug Repositioning - First-in-class Analysis - Competitive Analysis - Deals & Licensing

                  Schedule Your 30 min Free Demo!

"The FDA fast-track designation for TH1902 was supported by the data presented today. This designation is a significant recognition of our SORT1+ Technology as very few investigational therapies receive fast track designation based on preclinical data. It strongly endorses TH1902 as a potentially new and innovative treatment for all patients with sortilin positive (SORT1+) solid tumors that are refractory to standard therapy. The Phase 1 clinical trial is now underway and we look forward to advancing TH1902 through further stages of its development," said Dr. Christian Marsolais, Senior Vice President and Chief Medical Officer, Theratechnologies.

Mersana Therapeutics Presents Preclinical Data Highlighting Potential of XMT-2056 and XMT-1660 in Three Posters at Virtual 2021 AACR Annual Meeting

On April 10, 2021 Mersana Therapeutics, Inc. (NASDAQ:MRSN), a clinical-stage biopharmaceutical company focused on discovering and developing a pipeline of antibody-drug conjugates (ADCs) targeting cancers in areas of high unmet medical need, reported preclinical data from XMT-1660, a B7-H4-targeted Dolasynthen antibody-drug conjugate (ADC), and XMT-2056, an Immunosynthen-based STING-agonist ADC at the Virtual 2021 American Association for Cancer Research (AACR) (Free AACR Whitepaper) Annual Meeting being held from April 10-15th (Press release, Mersana Therapeutics, APR 10, 2021, View Source [SID1234577819]).

Schedule your 30 min Free 1stOncology Demo!
Discover why more than 1,500 members use 1stOncology™ to excel in:

Early/Late Stage Pipeline Development - Target Scouting - Clinical Biomarkers - Indication Selection & Expansion - BD&L Contacts - Conference Reports - Combinatorial Drug Settings - Companion Diagnostics - Drug Repositioning - First-in-class Analysis - Competitive Analysis - Deals & Licensing

                  Schedule Your 30 min Free Demo!

"The ability of Immunosynthen-based ADCs to activate the innate immune system via STING in tumor cells in addition to tumor-resident immune cells in a targeted manner could offer a significant therapeutic advantage over ADCs that modulate other immune activating pathways. These data demonstrate that XMT-2056 is highly differentiated from other innate immune activating approaches and has the anti-tumor activity and tolerability to support continued development of this novel STING-agonist ADC candidate," said Timothy B. Lowinger, Ph.D., Chief Science and Technology Officer of Mersana Therapeutics. "Additionally, we presented data showing that XMT-1660 outperformed other B7-H4 ADCs in vivo. The inversely correlated expression of B7-H4 and PD-L1 in breast tumors suggests an opportunity for a B7-H4 Dolasynthen ADC to address patients poorly served by checkpoint inhibitors. We expect to complete IND-enabling studies and advance both XMT-1660 and XMT-2056 into the clinic in early 2022."

"These encouraging data for both the Dolasynthen and Immunosynthen platforms demonstrate the scientific prowess of the Mersana research team and our commitment to discover and develop life-changing antibody-drug conjugates for patients fighting cancer," said Anna Protopapas, President and Chief Executive Officer of Mersana Therapeutics.

Details of the posters are as follows:

Poster Title: XMT-1660, a B7-H4-targeted Dolasynthen antibody-drug conjugate for the treatment of breast cancer
Poster Number: 907
Session Category: Experimental and Molecular Therapeutics
Session Title: Antibody Technologies

These data show that B7-H4 is a promising target for a Dolasynthen ADC due to its expression and function. B7-H4 is expressed across multiple different tumor types with high unmet medical need, including breast, endometrial and ovarian. XMT-1660 demonstrated robust in vivo activity against multiple triple-negative breast cancer models, as well as an ER+/HER2- breast cancer model, all of which express B7-H4.

In the MX-1 triple-negative breast model, XMT-1660 showed complete, durable regressions of tumors at a DolaLock payload dose of 0.15 mg/kg. In contrast, the DAR-2 and DAR-12 ADCs required twice the payload dose for comparable efficacy. XMT-1660 also showed superior efficacy at matched payload doses in the TNBC patient-derived xenograft model HBCx-24, and in the ER+/HER2- breast cancer PDX model HBCx-19 versus comparators.
Pharmacokinetics of XMT-1660 as well as the Dolasynthen DAR-2 and Dolaflexin DAR-12 comparator ADCs were evaluated in tumor-bearing mice and all were shown to be highly stable in vivo. Pharmacokinetics and tolerability of XMT-1660 and the Dolasynthen DAR-2 ADC were evaluated in non-human primates at equivalent payload doses. The PK and tolerability profiles were comparable and both ADCs exhibited high stability. These results, together with the superior efficacy of XMT-1660, support the selection of XMT-1660 for further development and for clinical study for the treatment of B7-H4-expressing tumors, such as breast, endometrial and ovarian.
Poster Title: XMT-2056, a well-tolerated, Immunosynthen-based STING-agonist antibody-drug conjugate which induces anti-tumor immune activity
Poster Number: 1738
Session Category: Immunology
Session Title: Immunomodulatory Agents and Interventions

These data suggest that XMT-2056, an Immunosynthen STING-agonist ADC, can overcome the limitations of the current therapeutic approaches, enabling tumor-targeted delivery of a STING agonist with improved efficacy and tolerability over a free IV STING agonist. Anti-tumor activity of Immunosynthen STING-agonist ADCs involves targeted activation of the STING pathway in both tumor-resident immune cells and tumor cells, delivering a one-two punch with the potential to increase the therapeutic index.

In vitro studies show that XMT-2056 has potent STING activity with >100-fold improvement in activity in comparison to the free STING-agonist payload.
XMT-2056 shows excellent in vivo efficacy even after a single IV dose, while having minimal effect on systemic cytokines. A single, low dose administration of XMT-2056 led to sustained tumor regressions in mice in comparison to the IV STING agonist which showed modest activity even at a dose approximately 100 times higher than that of the ADC. In contrast, when comparing the effect on systemic cytokine levels, the IV STING agonist had significantly higher levels compared to the STING-agonist ADC, which supports the hypothesis that a STING-agonist ADC can target STING activation to the tumor microenvironment, leading to improved anti-tumor activity and a significantly greater therapeutic index.
In vitro and in vivo studies demonstrate that STING agonist ADCs are able to activate the STING pathway in both tumor-resident immune cells and tumor cells, offering a potential advantage over other innate immune activating pathways.
To evaluate the safety profile, XMT-2056 was administered intravenously to non-human primates (NHP) in single and repeat-dose studies at multiple dose levels. XMT-2056 shows favorable pharmacokinetics in NHPs and is well tolerated at a dose level >10-fold higher than required for sustained tumor regression in mice models. Together these data support the clinical development of XMT-2056.
Poster Title: Tumor cell-intrinsic STING pathway activation leads to robust induction of Type III Interferons and contributes to the anti-tumor activity elicited by STING agonism
Poster Number: 1773
Session Category: Immunology
Session Title: Innate Immunity to Tumors

STING pathway agonism induces anti-tumor immunity by upregulating a Type I interferon response within the tumor microenvironment. While systemically or intra-tumorally administered free STING agonists are currently being evaluated in the clinic, these data suggest that a STING-agonist ADC, in which the STING agonist is conjugated to an antibody directed to a tumor antigen, can overcome the limitations of the current therapeutic approaches.

In vitro studies show that while most cancer cell lines do not respond to STING agonism in standard monoculture conditions, Immunosynthen STING-agonist ADCs do activate STING in the same cancer cells in the presence of immune cell-conditioned media, suggesting that the tumor cell-intrinsic STING pathway can be activated in the presence of cues from immune cells.
Nanostring analysis of human tumor xenografts reveal tumor cell specific induction of type III interferons (IFNs) by tumor cell-targeting Immunosynthen STING-agonist ADCs. In vitro studies confirmed the Type III interferon induction at the mRNA and cytokine level. Type III interferon production was markedly reduced in STING knock out cancer cell and immune cell co-cultures, suggesting that the tumor intrinsic STING activation is required for a robust Type III interferon induction in response to STING agonism. In addition, these data show that blocking Type III IFNs with neutralizing antibodies in cancer cell:immune cell co-cultures inhibits the production of key cytokines and cancer cell killing induced by STING-agonist ADC treatment, pointing to a potentially important role for Type III IFNs in anti-tumor immune responses downstream of STING pathway activation in tumor cells.
Together these data demonstrate that tumor cell intrinsic STING activation leads to a robust type III interferon induction, which contributes to the anti-tumor activity of tumor cell-targeted STING-agonist ADCs. This study supports the further development of Immunosynthen STING-agonist ADC candidates.

Positive Findings From Phase 1b Trial of GlycoMimetics’ GMI-1359 To Be Presented at AACR 2021 Meeting

On April 10, 2021 GlycoMimetics, Inc. (Nasdaq: GLYC) reported that a Phase 1b trial of GMI-1359, being conducted at Duke University Cancer Center, showed evidence of on-target effects, immune-activation and cell mobilization in the initial two patients treated with the Company’s dual antagonist of E-selectin and CXCR4 (Press release, GlycoMimetics, APR 10, 2021, View Source [SID1234577818]). Dorothy Sipkins, MD, PhD, Associate Research Professor in Pharmacology and Cancer Biology at Duke University School of Medicine, will present results from the proof-of-concept clinical study as well as a separate preclinical study supporting the positive biologic findings of the Phase 1b study. The presentation will be made at the American Association of Cancer Research (AACR) (Free AACR Whitepaper) 2021 Annual Meeting, which is being held virtually on April 10-15 and May 17-21. GMI-1359 is GlycoMimetics’ novel small molecule drug candidate, a dual antagonist of E-selectin and CXCR4, designed to target tumor-microenvironment resistance to chemotherapy in cancers with bone metastases.

Schedule your 30 min Free 1stOncology Demo!
Discover why more than 1,500 members use 1stOncology™ to excel in:

Early/Late Stage Pipeline Development - Target Scouting - Clinical Biomarkers - Indication Selection & Expansion - BD&L Contacts - Conference Reports - Combinatorial Drug Settings - Companion Diagnostics - Drug Repositioning - First-in-class Analysis - Competitive Analysis - Deals & Licensing

                  Schedule Your 30 min Free Demo!

The initial data from the study confirmed the dual CXCR4 and E-selectin antagonist’s on-target effects. In the two patients who completed treatment, evaluations of peripheral blood showed a consistent mobilization of CD34+ hematopoietic stem and progenitor cells at doses beginning at 5 mg/kg and a reduction of plasma levels of soluble E-selectin. Furthermore, in one individual, following the administration of 7.0 mg/kg of GMI-1359, an immunophenotyping assessment of peripheral blood showed a redistribution of myeloid derived suppressor cells (MDSCs) as evidenced by increased percentages of both the monocytic and granulocytic MDSCs. In this same individual following administration of 7.0 mg/kg GMI-1359, the incidence of M1 proinflammatory macrophages increased while the M2 anti-inflammatory macrophages, often associated with tumor progression, decreased. The clinical poster concludes that GMI-1359 demonstrated an acceptable safety and tolerability profile in the patients treated to date. No dose limiting toxicities were observed following multiple dose administration up to 7 mg/kg.

Dr. Sipkins noted, "Despite the fact that our patient numbers are very small due to COVID’s impact on recruitment, we are seeing the on-target effects of antagonizing both CXCR4 and E-selectin with use of GMI-1359, and that the drug is well-tolerated at all dose levels. Our pilot immune profile analysis also suggests that the drug could have favorable effects on the tumor immune microenvironment, echoing results seen in our preclinical work."

Dr. Sipkins will disclose preclinical evidence that it may be possible for GMI-1359 to augment immune recognition of the tumor. The data in the poster from a mouse metastatic breast carcinoma model demonstrated a reduction in the immune suppressive monocytic MDSCs at the primary tumor site and a significant increase in the CD8/Treg ratio in both the primary tumor and at the bone metastatic sites. These findings on immune cell redistributions strongly suggest the induction of a more favorable anti-tumor environment following GMI-1359 administration.

According to Dr. Eric J. Feldman, GlycoMimetics Senior Vice President and Chief Medical Officer, "The information shared in this AACR (Free AACR Whitepaper) poster provides us with important understandings upon which we expect to identify a potential indication for advancing GMI-1359 in the clinic. It suggests that this small molecule drug candidate could improve responses to therapies and potentially reduce the burden of metastatic breast cancer disease."

In prior preclinical research supported by GlycoMimetics, Dr. Sipkins’ laboratory demonstrated that E-selectin and CXCR4/SDF-1 interactions were critical for breast carcinoma cells (BCCs) invasion and retention, respectively, into bone. Moreover, they found that dormant and proliferating BCCs occupy distinct regions of the bone microenvironment, with dormant BCCs predominantly found in SDF-1 and E-selectin rich regions. These dormant BCCs are expected to be highly susceptible to GMI-1359 mobilization, suggesting a new intervention to break the foothold of dormant BC micrometastases in bone.

Details on the GMI-1359 e-presentation at the AACR (Free AACR Whitepaper) Meeting are as follows:

Title: Development of GMI-1359, a novel agent targeting tumor-microenvironment cross-talk in bone metastatic cancer
Presenter: Dorothy Sipkins, MD, PhD, Associate Research Professor in Pharmacology and Cancer Biology at Duke University School of Medicine
Session: e-Presentation
Date and Time: Saturday, April 10, 2021 (available online through Monday, June 21)

About GMI-1359

GMI-1359 is designed to simultaneously inhibit both E-selectin and CXCR4, which are adhesion molecules involved in tumor trafficking and metastatic spread. Preclinical studies indicate that targeting both E-selectin and CXCR4 with a single compound could improve efficacy in the treatment of cancers that involve the bone marrow, such as AML and multiple myeloma, or in solid tumors that metastasize to the bone, such as prostate cancer and breast cancer, as well as in osteosarcoma, a rare pediatric tumor affecting about 900 adolescents a year in the United States. GMI-1359 completed a Phase 1 clinical trial in healthy volunteers, and a Phase 1b clinical study is underway in breast cancer patients and is designed to enable investigators to identify study dose ranging and to generate initial biomarker data around the drug’s activity. In the first two patients evaluated, the study showed evidence of on-target effects, immune-activation and cell mobilization. GMI-1359 has received Orphan Drug designation and Rare Pediatric Disease designation from the FDA for the treatment of osteosarcoma.