false000126210400012621042024-05-212024-05-21

 

UNITED STATES
SECURITIES AND EXCHANGE COMMISSION
WASHINGTON, D.C. 20549

 

FORM 8-K

 

CURRENT REPORT

Pursuant to Section 13 or 15(d) of the Securities Exchange Act of 1934

Date of Report (Date of earliest event reported): May 21, 2024

 

 

MEI Pharma, Inc.

(Exact name of Registrant as Specified in Its Charter)

 

 

Delaware

001-41827

51-0407811

(State or Other Jurisdiction
of Incorporation)

(Commission File Number)

(IRS Employer
Identification No.)

 

 

 

 

 

11455 El Camino Real, Suite 250

 

San Diego, California

 

92130

(Address of Principal Executive Offices)

 

(Zip Code)

 

Registrant’s Telephone Number, Including Area Code: 858 369-7100

 

 

(Former Name or Former Address, if Changed Since Last Report)

 

Check the appropriate box below if the Form 8-K filing is intended to simultaneously satisfy the filing obligation of the registrant under any of the following provisions:

Written communications pursuant to Rule 425 under the Securities Act (17 CFR 230.425)
Soliciting material pursuant to Rule 14a-12 under the Exchange Act (17 CFR 240.14a-12)
Pre-commencement communications pursuant to Rule 14d-2(b) under the Exchange Act (17 CFR 240.14d-2(b))
Pre-commencement communications pursuant to Rule 13e-4(c) under the Exchange Act (17 CFR 240.13e-4(c))

Securities registered pursuant to Section 12(b) of the Act:


Title of each class

 

Trading
Symbol(s)

 


Name of each exchange on which registered

Common Stock, $0.00000002 par value

 

MEIP

 

The Nasdaq Stock Market LLC

Indicate by check mark whether the registrant is an emerging growth company as defined in Rule 405 of the Securities Act of 1933 (§ 230.405 of this chapter) or Rule 12b-2 of the Securities Exchange Act of 1934 (§ 240.12b-2 of this chapter).

Emerging growth company

If an emerging growth company, indicate by check mark if the registrant has elected not to use the extended transition period for complying with any new or revised financial accounting standards provided pursuant to Section 13(a) of the Exchange Act.

 


Item 7.01 Regulation FD Disclosure.

On May 21, 2024, Richard Ghalie, M.D., the Chief Medical Officer of MEI Pharma, Inc., will participate in a panel at the Brookline Virtual MDS and AML Symposium. During the panel discussion Dr. Ghalie will discuss clinical data from the ongoing clinical evaluation of voruciclib in patients with Acute Myeloid Leukemia, which are included in the presentation attached as Exhibit 99.1 to this Form 8-K and are incorporated by reference into this Item 7.01.

The information in this Form 8-K and the Exhibit attached hereto shall not be deemed “filed” for purposes of Section 18 of the Securities Exchange Act of 1934, as amended, nor shall it be deemed incorporated by reference in any filing under the Securities Exchange Act of 1933, as amended, except as shall be expressly set forth by specific reference in such filing.

 

Item 9.01 Financial Statements and Exhibits.

 

 

 

Exhibit

No.

 

Description

 

 

99.1

 

MEI Pharma, Inc., Corporate Presentation May 21, 2024

 

 

104

 

Cover Page Interactive Data File – the cover page interactive data file does not appear in the Interactive Data File because its XBRL tags are embedded within the XBRL document.

 


SIGNATURES

Pursuant to the requirements of the Securities Exchange Act of 1934, the registrant has duly caused this report to be signed on its behalf by the undersigned hereunto duly authorized.

 

 

 

MEI Phama, Inc.

 

 

 

 

Date:

May 21, 2024

By:

/s/ David M. Urso

 

 

 

David M. Urso
Chief Executive Officer and Director

 


Slide 1

NASDAQ: MEIP Corporate Overview Novel drug candidates to address known resistance mechanisms to standard-of-care cancer therapies May 21, 2024


Slide 2

Forward Looking Statements Certain information contained in this communication that are not historical in nature are "forward-looking statements" within the meaning of the "safe harbor" provisions of the Private Securities Litigation Reform Act of 1995 including, without limitation, statements regarding: the potential, safety, efficacy, and regulatory and clinical progress of our product candidates, including the anticipated timing for initiation of clinical trials and release of clinical trial data and our expectations surrounding potential regulatory submissions, approvals and timing thereof, our business strategy and plans and the sufficiency of our cash, cash equivalents and short-term investments to fund our operations. You should be aware that our actual results could differ materially from those contained in the forward-looking statements, which are based on management's current expectations and are subject to a number of risks and uncertainties, including, but not limited to, risk relating to our ability to successfully commercialize our product candidates; the availability or appropriateness of utilizing the FDA’s accelerated approval pathway for our product candidates; final data from our pre-clinical studies and completed clinical trials potentially differing materially from reported interim data from ongoing studies and trials; costs and delays in the development and/ or FDA approval, or the failure to obtain such approval, of our product candidates; uncertainties or differences in interpretation in clinical trial results; uncertainty regarding the impact of rising inflation and the increase in interest rates as a result; potential economic downturn; activist investors; our inability to maintain or enter into, and the risks resulting from, our dependence upon collaboration or contractual arrangements necessary for the development, manufacture, commercialization, marketing, sales and distribution of any products; competitive factors; our inability to protect our patents or proprietary rights and obtain necessary rights to third party patents and intellectual property to operate our business; our inability to operate our business without infringing the patents and proprietary rights of others; general economic conditions; the failure of any products to gain market acceptance; our inability to obtain any additional required financing; technological changes; government regulation; changes in industry practice; and one-time events. We do not intend to update any of these factors or to publicly announce the results of any revisions to these forward-looking statements. Under U.S. law, a new drug cannot be marketed until it has been investigated in clinical studies and approved by the FDA as being safe and effective for the intended use.


Slide 3

Clinical-stage Oncology Company Evaluating Novel Drug Candidates to Address Known Resistance Mechanisms to Standard-of-care Cancer Therapies Significant Addressable Markets Near-term Value Drivers Clinical Data Support Combinations Cash sufficient to fund operations for at least the next 12 months Two Novel Drug Candidates 3 Science Driven Development of Combination Therapies


Slide 4

Two Novel Oncology Drug Candidates Investigational Agents Therapeutic Area Combination Pre-IND Phase 1/1b Phase 1/2 Voruciclib Oral CDK9 Inhibitor Acute Myeloid Leukemia Relapsed/refractory (2L+) Single-agent VENCLEXTA® (venetoclax) Solid Tumors* Single-agent & Vemurafenib  ME-344 OXPHOS Inhibition HER2-negative Breast Cancer** AVASTIN® (Bevacizumab) Colorectal Cancer Relapsed AVASTIN® (Bevacizumab) Solid Tumors VEGF Inhibitors (Bevacizumab & tyrosine kinase inhibitors) Completed Ongoing *Three Phase 1 Studies in a total of 77 patients. **Phase 0 window of opportunity study: investigator initiated; placebo controlled. Completed Completed New Formulation VORUCICLIB: Oral CDK9 Inhibitor Plan to initiate Phase 2 arm in R/R AML 2025 Encouraging results in AML patients treated, including 42 in combination with venetoclax (Venclexta®) Mutation agnostic therapy: potential to address larger % of AML population than menin, FLT3 or IDH inhibitors ME-344: Novel Inhibitor of OXPHOS Clinical data support combination with bevacizumab (Avastin®) in metastatic colorectal cancer Novel approach intended to address cancers where VEGF inhibition is SoC Readying new formulation for Phase 1 Completed


Slide 5

Voruciclib: A Selective Oral CDK9 Inhibitor Drug Candidate


Slide 6

Voruciclib: A Selective Oral CDK9 Inhibitor with Potential to Address Hematologic Malignancies and Solid Tumors Dey, et al. Sci Reports (2017) 7(1):8007. Luedtke, et al. Signal Transduct Ther (2020) 5(1):17. Wiley, et al. Cancer Research (2021) 81(13 supp):1962 6 Myeloid leukemia 1 (MCL-1): An antiapoptotic protein of the BCL-2 family that prevents apoptosis by binding to the pro-apoptotic BCL-2 proteins. Myc proto-oncogene: A transcription factor that promotes cell growth and proliferation and regulates cell metabolism. Voruciclib Inhibits CDK9 Leading to Decreased Mcl-1 and Myc Proteins Voruciclib is Differentiated Oral administration Favorable PK: Half-life of 26-32 hours allows once a day dosing Selective Higher specificity and longer residence time on target vs CDK 6, 4 &1 Greater selectivity against CDKs relative to other kinases Potent IC50 from 0.2 to 1.7 μM in various cell lines


Slide 7

MCL-1: Increase associated with poor prognosis in AML and several B-cell malignancies Upregulation is an established venetoclax resistance mechanism Venetoclax inhibits BCL2 but can lead to stabilization of Mcl-1 MYC: Over expressed in many cancers, including those with KRAS mutations CDK9 inhibition leads to reduced transcription and stability of MYC Voruciclib inhibits MCL-1 and MYC via CDK9 inhibition 7 Both Mcl-1 and Myc Proteins Represent Important Therapeutic Targets for Hematologic Malignancies and Solid Tumors


Slide 8

Voruciclib Program Overview Significant clinical experience: Over 150 patients treated to date, 82 with hematological malignancies and 77 with solid tumors Current focus on combination with venetoclax in R/R AML: Addresses established venetoclax resistance mechanism High Need: Poor outcomes with current salvage therapies after venetoclax failure Phase 1 continues enrolling with additional combination readouts in H2 2024 Voruciclib is a Novel Oral CDK9 Inhibitor in Development for R/R AML in Combination with Venetoclax Planned Dose Optimization in H2 2024 Followed by Ph 2 arm at RP2D in 2025, Creating Robust Ph 3 Read Package: Over 100 pts with r/r AML, including ~85 administered V+V, ~40 at RP2D (Phase 2 study arm subject to FDA advice)


Slide 9

Poor Outcomes Post Venetoclax Failure in Patients with No Actionable Mutations Maiti, A., et. al., Haematologica 2021: 106(3. Maiti, A., et. al.,  Blood 2023: 142 (Supplement 1): 4289. Zainaldin, C., et. al., Leuk Lymphoma 2022: 63(13):3245-3248. Daver, N., Blood (2022) 140 (Supplement 1): 141–144: 9 * Includes: intensive chemo based, non-intensive chemo based, single agent targeted therapy, immunotherapy, Allo-SCT Various Regimens* Venetoclax +decitabine/ cedazuridine IC, targeted therapy, clinical trial Ven+AZA +Magro CR+CRh/CRi in Venetoclax Exposed R/R AML Patients1 Achieving a Complete Response Rate of ≥20% with VORU+VEN in R/R AML Would Represent a Significant Improvement Over Current Options CR+CRh/CRi Rates for Non-mutation Targeted Regimens Range from 6-12.5% in Patients Previously Exposed to Venetoclax


Slide 10

High Unmet Need Remains for ≥50% of AML Population With No Actionable Mutations 10 1. Miyamoto, K., et. al., Int J Mol Sci. 2020;21(14):5114. 2. Malani, D., Cancer Discov. 2022;12(2):388-401 Diagram is for illustrative purposes. In comparison, approximately 30%, 30% and 20% of AML patients have NPM1, FLT3 and IDH1/2 mutations, respectively1. In one study, approximately 80% of patients with an NPM1 mutation had a co-mutation in either FLT3, IDH1/2 or both2 High Need For Mutation Agnostic Therapy: There is significant overlap across mutation sub-populations1 50-55% of AML Patients Do Not Have Actionable Mutations1,2


Slide 11

Large Potential Addressable Population with High Unmet Need for Mutation Agnostic Therapy Venclexta sales were ~$2.3B in 2023 WW across all indications; projected to grow to ~$3.3B by 2028 Sources: Evaluate, April 2024; Clarivate November 2023 Use of Venetoclax Based Therapy Increasing Across AML Treatment in Various Combinations Venetoclax + HMA is entrenched as the dominant SOC in 1L unfit and expected to grow Ongoing studies expected to shift use of mutation targeted agents up to 1L in combination with Venetoclax + HMA backbone as triplet regimens Novel venetoclax combinations being studied in R/R AML R/R AML Treatable Annually (U.S.) R/R AML Treatable w/o Actionable Mutations Annually (U.S.) ~6K Patients ~12K Patients ≥50% w/ No Actionable Mutation Addressable population in R/R AML will grow as use of mutation targeted agents moves up to 1L ~6K Addressable Patient Population for Mutation Agnostic Therapy in Patients with R/R AML


Slide 12

Preclinical Studies Demonstrate Voruciclib Suppresses Mcl-1 and Synergizes with Venetoclax in AML Murine Xenograft Model Luedtke, et al. Signal Transduct Ther (2020) Extends Survival in MV4-11 Tumor c. Suppresses Mcl-1 Level Increases Apoptosis VOR (nM) VEN (nM) 0 0 0 12.5 0 25 0 50 500 0 1000 0 2000 0 1000 12.5 1000 25 2000 12.5 2000 25 VEN VOR VOR + VEN a. b. 12 Voruciclib also synergizes with venetoclax in CLL patients1,2 and in High Risk DLBCL Murine Models3 1. Lesnick et al. J Clin Oncol.2020;38(15_suppl):e20009; 2. Paiva et al, PLoS ONE. 2015;10(11):e0143685; 3. Dey et al. Nature Sci Rep. 2017; 7:18007


Slide 13

13 Plan to Initiate a Phase 2 Study Arm in 2025 After Determining Recommended Phase 2 Dose and Schedule (Phase 2 study arm subject to FDA advice) Ph 1/2 Study to Generate Data on ~106 Patients With R/R AML Administered Voruciclib Alone or with Venetoclax IS2w,2w: Intermittent dosing of voruciclib daily for 2 weeks in a 4-week cycle. IS3w,1w: Intermittent dosing of voruciclib daily for 3 weeks in a 4-week cycle. RP2D: Recommended Phase 2 Dose Study Population Relapsed/Refractory AML Relapsed/Refractory B-cell malignancies Dose escalation with standard 3+3 design Single agent Combination with venetoclax Endpoints Safety and tolerability Pharmacokinetics Biologic correlative studies BH3 profiling, MCL-1 expression Molecular mutation analysis Preliminary activity Ongoing Phase 1 Evaluation of Voruciclib + Venetoclax in R/R AML: (N = ~66 pts in dose escalation and expansion cohorts) IS2w,2w IS3w,1w RP2D & Schedule Dose Escalation N=29 50 mg to 300 mg N=12 150 mg to 300 mg Dose Expansion N=12 300 mg N=12 250 mg* Phase 2 Arm N=20 * Final dose determination to be based on data from escalation cohort.


Slide 14

Single-Agent Voruciclib Well-Tolerated, Showed Clinical and Biomarker Activity in AML & CLL AML CLL Davids et al, ASH 2023 14 Analysis of patient AML blasts showed a decrease in RNA Pol II phosphorylation after voruciclib treatment. CDK9 directly phosphorylates RNA Pol II. Longitudinal transcriptomic analysis of blood samples from CLL patients shows a trend towards reduced Mcl-1 mRNA. Voruciclib up to 200mg for 14 days in a 28-day Cycle Demonstrated Anti-Leukemic Activity . . . Single-Agent Correlative Studies Demonstrates on-Target Biologic Activity Single-agent activity (AML, n=21) at low dose studied in heavily pretreated patients: 1 Patient: MLFS (81 yo, 4 prior lines, adverse mutations & cytogenetics) 5 of 10 pts with AML at 200mg had stable disease 2 patients had differentiation syndrome demonstrative of biological activity Patient characteristics: 21 AML, 19 B-cell malignancies; Median prior therapies: 3 (range, 1-8) No dose-limiting toxicity* No drug-related neutropenia No Grade 3+ drug related toxicity No discontinuation due to drug related toxicity Dose escalation in Cohort 2 was stopped at 200 mg before reaching the MTD on this schedule to focus on evaluation of combination with venetoclax. . . . and Was Well-tolerated on Intermittent Dosing


Slide 15

Evidence of Activity and Tolerability in Dose Escalation Groups of Voruciclib Plus Venetoclax in R/R AML All V+V patients are heavily pretreated with a median of 2 prior therapies (range 1-7) including venetoclax in 28/29 patients 15 MEI Data on file 29 patients in dose escalation cohorts (IS2w,2w) Clinical activity observed: Responses Reduced transfusions Improved blast counts Mcl-1 decreases, including: A greater decrease at a higher dose Response related decreases No DLTs observed No significant increase in myelosuppression PK analysis does not show drug-drug interaction


Slide 16

Combination Arm Characteristics: Patients Typically Heavily Pretreated With a Median of 3 Prior Therapies and High Rate of Adverse Molecular and Cytogenetic Features 16   Total (N=29) 2017 ELN Risk Category Favorable Intermediate Adverse 3 (11%) 5 (17%) 21 (72%) Poor Cytogenetics Patients with adverse cytogenetics 14 (48%) Adverse Molecular Mutations Patients with adverse mutations TP53 ASLX1 RUNX1 GATA2   19 (66%) 7 (24%) 7 (24%) 4 (14%) 2 (7%) Baseline Bone Marrow Blast Median (range) 45% (2-77%) N = 29 Number of prior therapies Median (range) ≥3 prior 3 (1-7) 15 (52%) Prior HSCT 9 (31%) Prior venetoclax 1st line ≥2nd line 28 (97%) 14 (50%) 14 (50%) Prior anthracyclines 18 (62%) Median age 67 years (range 34-89) 15 (52%) had ≥3 prior lines 28/29 (97%) treated with venetoclax in earlier line of therapy 21 (72%) with adverse 2017 ELN Risk Category MEI Data on file


Slide 17

Voruciclib Dosed up to 300 mg Appeared Generally Well-Tolerated with No Apparent Dose Response to Adverse Events Reported 17   (50 mg QOD) (N=6) (50 mg QD) (N=3) (100 mg QD) (N=4) (150 mg QD) (N=4) (200 mg QD) (N=4) (250 mg QD) (N=4) (300 mg QD) (N=4) Total (N=29) Nausea 0 0 2 (50.0) 3 (75.0) 2 (50.0) 1 (25.0) 2 (50.0) 10 (34.5) Platelet Count Decreased 0 1 (33.3) 1 (25.0) 3 (75.0) 1 (25.0) 1 (25.0) 1 (25.0) 8 (27.6) Febrile Neutropenia 0 1 (33.3) 2 (50.0) 2 (50.0) 0 1 (25.0) 1 (25.0) 7 (24.1) Anaemia 0 0 2 (50.0) 2 (50.0) 0 1 (25.0) 1 (25.0) 6 (20.7) Hypokalaemia 0 0 2 (50.0) 1 (25.0) 2 (50.0) 1 (25.0) 0 6 (20.7) Cough 2 (33.3) 0 1 (25.0) 1 (25.0) 1 (25.0) 0 0 5 (17.2) Diarrhoea 1 (16.7) 0 1 (25.0) 1 (25.0) 1 (25.0) 1 (25.0) 0 5 (17.2) Dyspnoea 2 (33.3) 0 1 (25.0) 1 (25.0) 0 1 (25.0) 0 5 (17.2) Fatigue 0 0 0 3 (75.0) 1 (25.0) 0 1 (25.0) 5 (17.2) Stomatitis 2 (33.3) 0 1 (25.0) 1 (25.0) 0 0 0 4 (13.8) Vomiting 0 0 1 (25.0) 0 0 2 (50.0) 1 (25.0) 4 (13.8) Anxiety 1 (16.7) 0 2 (50.0) 0 0 0 0 3 (10.3) Corona Virus Infection 1 (16.7) 0 0 1 (25.0) 1 (25.0) 0 0 3 (10.3) Hypotension 1 (16.7) 0 1 (25.0) 1 (25.0) 0 0 0 3 (10.3) Ph 1 Study Treatment Emergent Adverse Events in ≥10% of Patients MEI Data on file


Slide 18

Responses and Blast Count Reductions Demonstrate Evidence of Anti-Leukemic Activity in Heavily Pretreated Patients After Venetoclax Failure 18 ≥100 50% of Evaluable Patients with Pre/Post Bone Marrow Had a Decrease in Blast 85% (17/20) of patients administered voruciclib at ≥100 mg achieved response or stable disease: 3 patients achieved a response 2 patients achieved a complete response with incomplete hematologic recovery (CRi) 1 patient achieved a morphologic leukemia-free state (MLFS) Each responding patient received venetoclax in an earlier line of treatment. Responses lasted 7 months in one patient, 5 months and ongoing in the second patient; the third patient was referred to stem cell transplant. 14 patients had stable disease, which lasted more than 90 days in 5 patients. Best Change in Bone Marrow Blast Count MEI Data on file


Slide 19

Decreases in Mcl-1 Demonstrate On-target Biological Activity Relative Mcl-1 Protein By Response MEI Data on file Relative Mcl-1 Protein in PBMC Greater Decrease in Mcl-1 Protein in Responders Mcl-1 Increases After Venetoclax Then Decreases with Voruciclib More Consistent Decrease in Mcl-1 at Higher Doses Preliminary analyses based on available patient samples Bars represents mean of Mcl-1 protein levels at various timepoints Mcl-1 protein expressed as mean fluorescence normalized to D1 0h or D3 0h Mcl-1 increases after venetoclax dosing (D1 0h – D3 0h) and decreases after 1st voruciclib dosing (D3 6h), continuing after 12 daily doses (D14 0h) Relative Mcl-1 Protein Abundance By Dose


Slide 20

24 patients had serial blood tests for blasts with values >0 18/24 pts (75%) had decreased balsts on Day 14, at the end of voruciclib dosing 8/18 pts (44%) had increased blasts between Day 14 and 28, when off voruciclib but receiving venetoclax Peripheral Blast Counts Decrease on Voruciclib + Venetoclax Combination (Days 1 to 14) and Rebound with Venetoclax Alone (Days 15-28) 20 Ongoing Phase 1 Study Evaluating Voruciclib administered on days 1 to 21 of the 28-Day Cycle to Extend Voruciclib Exposure MEI Data on file


Slide 21

Near-term Data to Inform RP2D for Phase 2 Study Arm in R/R AML Anticipated Total of ~40 Patients at RP2D & Schedule to Support Phase 3 Ready Program Potential H2 2024 Updates: IS2w,2w Dose Expansion (N=12) IS3w,1w Dose Escalation/Expansion (N=12/12) Phase 2 Study Arm* – Initiation and Data in 2025 RP2D and Schedule: N=~20 *Phase 2 study arm subject to FDA advice


Slide 22

Voruciclib Presents Novel Mechanism to Evaluate Treating a Range of Solid Tumors via MYC Inhibition


Slide 23

Opportunities to Address both Hematologic Malignancies and Solid Tumors MCL-1: Increase associated with poor prognosis in AML and several B-cell malignancies Upregulation is an established venetoclax resistance mechanism Venetoclax inhibits BCL2 but can lead to stabilization of Mcl-1 MYC: Over expressed in many cancers, including those with KRAS mutations CDK9 inhibition leads to reduced transcription and stability of Myc Voruciclib inhibits MCL-1 and MYC via CDK9 inhibition 23 Voruciclib is a Selective Oral CDK9 Inhibitor Targeting Cell Proliferation Regulation


Slide 24

2 weeks on, 1 week off schedule (N = 29 pts) 75 to 850 mg MTD = 600 mg 41% disease control rate Daily continuously schedule (N = 39 pts) 75 to 500 mg MTD = 350 mg 31% disease control rate Safety data No evidence of myelosuppression Most common AEs involved GI tract Single-Agent Phase 1 Studies in Solid Tumors Demonstrated Reduction in MYC and was Generally Well-tolerated at Expected Therapeutic Doses 10 gene biomarkers evaluated in blood in daily dosing study c-MYC expression decreased in ~60% patients tested (n=25) 24 Gupta et al, ASCO 2012; Hao et al, ASCO 2012; MEI Data on file


Slide 25

CDK9 can influence MYC protein stability in KRAS mutant cancer cells Cell Stimulatory Signals KRAS PI3K GSK3β CDK9 RAF MEK MYC MYC MYC protein degradation by proteasome SCF-Fbw7 Ubiquitin sotorasib adagrasib pT58 Pin1 Pin1 PP2A pS62 pS62 pT58 (unstable) Voruciclib (stable) Transcriptional activation of MYC target genes MYC MYC ERK1 CDK9 Mutations in KRAS at G12, G13, and Q61 are oncogenic drivers in many cancers, including lung, colorectal, pancreatic, bone marrow, and endometrial carcinomas. KRAS mutations are frequently accompanied by stabilization of the MYC oncoprotein through increased MYC transcription and decreased protein degradation. MYC protein stability is mediated by phosphorylation of MYC on Ser 62 by ERK and CDK9 kinases. Wiley, SE. et al. AACR, 2021. 25


Slide 26

VOR inhibited proliferation of KRAS mutant cell lines from multiple indications CRC Esophageal Multiple Myeloma NSCLC Ovarian PDAC VOR inhibited proliferation of cell lines with various KRAS mutations G12C, G12D, G12A, G12V G13C, G13D Q61H Wiley, SE. et al. AACR, 2021 & unpublished. Time (days) Ave tumor weight (mg) 600 500 400 100 200 300 0 0 2 4 6 8 10 12 14 1800 1600 1400 1200 1000 800 600 400 200 0 0 2 4 6 Time (days) Ave tumor weight (mg) 8 10 12 Control VOR 100 mg/kg VOR 50 mg/kg VOR 200 mg/kg 6000 5000 4000 3000 2000 1000 0 0 2 4 6 Time (days) Ave tumor weight (mg) 8 10 12 Control VOR 100 mg/kg VOR 50 mg/kg VOR 200 mg/kg Control VOR 100 mg/kg VOR 200 mg/kg HCT-116 (CRC, KRAS G13D) SW-480 (CRC, KRAS G12V) H460 (NSCLC, KRAS Q61H) Voruciclib Inhibits KRAS Mutant Cell Lines In Vitro and In Vivo in Xenograft Mice 26


Slide 27

Combining Voruciclib with Sotorasib Results in Enhanced Cell Death in an in vivo MIA PaCa-2 Tumor Model Con VOR SOTO Combo DAPI 40X Cell death around each microinjection site measured by nuclear condensation and fragmentation 27 Wiley, SE. et al. AACR, 2021.


Slide 28

ME-344: An OXPHOS Inhibitor Drug Candidate


Slide 29

ME-344: A Novel Inhibitor of OXPHOS with Dual Effect on OXPHOS and Purine Synthesis 1 MEI data on file, 2Lim et al, Am J Cancer Res. 2015, 3Manevich et al, J Pharm and Exper Ther. 2016, 4Hurrish et al, Biochem Pharmacol 2024 Potent Inhibitor of Proliferation nM potency against majority of 240 cell lines from solid tumors and hematologic malignancies1 Inhibition of OXPHOS leads to Reduced mitochondrial membrane potential, increased ROS, and decreased ATP2,3 Induced cell death through multiple signaling pathways Caspase and non-caspase mediated DNA fragmentation Increased AMPK signaling leading to decreased mTOR signaling and destructive autophagy Reduction in de novo Purine Biosynthesis Decreased de novo purine biosynthesis, an important pathway in cancer cells4 29


Slide 30

Novel MOA with OXPHOS inhibition leading to ATP depletion in malignant cells Significant need in large addressable patient populations: anti-angiogenics like bevacizumab and TKIs used in multiple settings as SOC, including: Colorectal, ovarian, renal cell, glioblastoma and pancreatic cancers Clinical data set supporting activity in combination with bevacizumab in a well-tolerated manner, including: Phase 1b study in patients with mCRC and controlled Phase 0 study in HER2 negative breast cancer Current program focuses developing new formulation with potential to increase biological activity, patient convenience and commercial opportunity ME-344 Program Overview: Inducing Synthetic Lethality in Solid Tumors in Combination with Anti-angiogenics


Slide 31

ME-344 blocks the production of ATP by inhibiting the OXPHOS pathway Anti-angiogenic therapies reduce glycolysis Cancer cells switch between mitochondria and glycolytic metabolic pathways to escape the blocking of either energy source The potential to inhibit both mitochondrial energy production via ME-344 and glycolytic energy production via VEGF inhibition is intended to result in metabolic synthetic lethality of cancer cells 3 Energy for cancer cell growth 2 Glycolytic Energy 1 Mitochondrial Energy 4 Tumor cell proliferation and growth ME-344 Avastin X X ME-344: A Novel Combination Approach with Anti-angiogenic Therapies Like Avastin® to Potentially Address Multiple Cancers 31


Slide 32

ME-344 + Regorafenib in CRC Model2 Survival ME-344 Synergizes with Anti-angiogenics in Multiple Murine Solid Tumor Models Resulting in Tumor Growth Delay and Improved Survival 32 1 Navarro et al, Cell Reports 2016, 2 MEI Data on file ME-344 + TKI in PyMT Breast Cancer Model1 Tumor Volume OXPHOS Reduced tumor growth and OXPHOS Tumor Growth Delay Stivarga (60%), ME-344 (4%), Stivarga + ME-344 (96%) ME-344 + Nintedanib in Lung Cancer Models2 Pulm 24 NSCLC: Tumor Volume EBC1 Squamous cell: Tumor Volume Reduced tumor growth Reduced tumor growth, extended survival


Slide 33

Clinical Study Objectives Assess ability of bevacizumab to shift tumor reliance from glycolysis to mitochondrial metabolism Assess ability of ME-344 + Avastin to inhibit tumor proliferation compared to Avastin + placebo Arm A N = 21 Bevacizumab 15 mg/kg day 1 ME-344 10mg/kg days 8, 15, 21 Arm B N = 20 Bevacizumab 15 mg/kg day 1 Saline days 8, 15, 21 Analysis: FDG-PET: days 1 and 28 Biopsy: days 1 and 28 Sponsored by Spanish National Cancer Research Centre Qunitela-Fandino, Clin Cancer Res (2020) 26 (1): 35–45. Establishing Proof of Concept: ME-344 in Combination with Bevacizumab Demonstrates Ki67 Decrease Supporting Novel Therapeutic Strategy to Induce Synthetic Lethality in Tumors 33 Randomized, Controlled Phase 0 Study ME-344 in combination with bevacizumab in treatment-naïve HER2-negative breast cancer patients demonstrated biologic activity as evidenced by a decrease of proliferation biomarker Ki67. ME-344 + Bevacizumab Saline + Bevacizumab


Slide 34

Patients with mCRC disease after failure of standard therapies Primary objective: 16-week PFS Secondary objectives: ORR, OS, safety Continue therapy until disease progression or toxicity Cohort 2 (not enrolled) Option for second 20 patient Cohort 2 if PFS at 4-mo ≥20% in Cohort 34 Cohort 1 (N=23) ME-344 at 10 mg/kg Day 1,8, 15 Bevacizumab 5 mg/kg Day 1, 15 28-day cycle Clinical Support of Concept: Phase 1B Evaluating ME-344 Plus Bevacizumab in Patients with Relapsed Metastatic Colorectal Cancer Cohort 1: Heavily pretreated patient population Median of 4 prior therapies (range 1-8) 100% of patients had prior chemotherapy 100% of patients had prior bevacizumab 39% had prior Lonsurf Tumor characteristics at diagnosis: R & transverse colon: 26% L colon: 43% Rectum: 31% Liver metastases at enrollment: 70% MEI Data on file


Slide 35

Phase 1b Further Supports ME-344 Novel Therapeutic Strategy to Induce Synthetic Lethality 5 of 20 (25%) evaluable patients completed 16 weeks of therapy without evidence of disease progression, exceeding the 20% predetermined threshold as set forth in the Clinical Study Protocol. No overlapping toxicities observed. 2 (9%) patients discontinued therapy due to AE: fatigue considered related to study drugs and sepsis considered unrelated. Most common (≥10%) drug-related adverse events (all grades/grade ≥3): Fatigue in 8 (35%) / 3 (13%). Abdominal pain in 3 (13%) / 2 (9%) patients. Cohort 1 Exceeds Predetermined Non-progression Threshold Generally Well-tolerated MEI Data on file


Slide 36

Phase 1b: Encouraging Progression Free Survival and Overall Survival No. (%) of patients (N = 23) Complete Response 0 Partial Response 0 Stable Disease 9 (39.1%) Progressive Disease 11 (47.8%) Not Evaluable for response 3 (13.0%) Median PFS = 1.9 m (95% CI: 1.6-5.2) 4 m PFS = 31.2% (95% CI: 13.5-50.7) 15 March 2024 Data cutoff Median OS = 6.7 m (95% CI: 3.4-NA) 15 March 2024 Data cutoff MEI Data on file


Slide 37

All Adverse Events in ≥3 Patients   All Subjects (N=23)    N (%) Grade 1 Grade 2 Grade 3 Grade 4 Grade 5 All Grade Fatigue 4 (17.4) 2 (8.7) 3 (13.0) 0 0 9 (39.1) Abdominal pain 3 (13.0) 3 (13.0) 2 (8.7) 0 0 8 (34.8) Diarrhea 2 (8.7) 4 (17.4) 1 (4.3) 0 0 7 (30.4) Constipation 4 (17.4) 1 (4.3) 0 0 0 5 (21.7) Blood sodium decreased 2 (8.7) 2 (8.7) 0 0 0 4 (17.4) Nausea 2 (8.7) 1 (4.3) 1 (4.3) 0 0 4 (17.4) Vomiting 2 (8.7) 1 (4.3) 1 (4.3) 0 0 4 (17.4) Blood bilirubin increased 1 (4.3) 0 2 (8.7) 0 0 3 (13.0) Dehydration 0 0 3 (13.0) 0 0 3 (13.0) Hypertension 0 1 (4.3) 2 (8.7) 0 0 3 (13.0) Leukocytosis 0 0 3 (13.0) 0 0 3 (13.0) Non-cardiac chest pain 3 (13.0) 0 0 0 0 3 (13.0) MEI Data on file


Slide 38

ME-344 to Continue Advancing via Development of a New Formulation ME-344 New Formulation with Potential to: Increase Biological Activity Improve Patient Convenience Enhance Commercial Opportunity New Formulation Update Expected H1 2025


Slide 39

NASDAQ: MEIP Financial Overview


Slide 40

Financial Highlights As of December 31, 2023, MEI had $59.5 million in cash, cash equivalents, and short-term investments with no outstanding debt. The Company believes its cash balance is sufficient to fund operations for at least the next 12 months


Slide 41

NASDAQ: MEIP Corporate Overview Novel drug candidates to address known resistance mechanisms to standard-of-care cancer therapies May 21, 2024

v3.24.1.1.u2
Document And Entity Information
May 21, 2024
Cover [Abstract]  
Document Type 8-K
Amendment Flag false
Document Period End Date May 21, 2024
Entity Registrant Name MEI Pharma, Inc.
Entity Central Index Key 0001262104
Entity Emerging Growth Company false
Entity File Number 001-41827
Entity Incorporation, State or Country Code DE
Entity Tax Identification Number 51-0407811
Entity Address, Address Line One 11455 El Camino Real, Suite 250
Entity Address, City or Town San Diego
Entity Address, State or Province CA
Entity Address, Postal Zip Code 92130
City Area Code 858
Local Phone Number 369-7100
Written Communications false
Soliciting Material false
Pre-commencement Tender Offer false
Pre-commencement Issuer Tender Offer false
Title of 12(b) Security Common Stock, $0.00000002 par value
Trading Symbol MEIP
Security Exchange Name NASDAQ

MEI Pharma (NASDAQ:MEIP)
過去 株価チャート
から 5 2024 まで 6 2024 MEI Pharmaのチャートをもっと見るにはこちらをクリック
MEI Pharma (NASDAQ:MEIP)
過去 株価チャート
から 6 2023 まで 6 2024 MEI Pharmaのチャートをもっと見るにはこちらをクリック