Attached files

file filename
EX-99.1 - PRESS RELEASE - LUMOS PHARMA, INC.nlnk-20180416x8kxex991.htm
8-K - 8-K - LUMOS PHARMA, INC.nlnk-20180416x8k.htm
American Association of Cancer Research (AACR) 2018 Theodore S. Johnson, MD, PhD Georgia Cancer Center – Augusta University April 15, 2018 Front-line Therapy of DIPG Using IDO Pathway Inhibitor Indoximod in Combination with Radiation and Chemotherapy


 
Cautionary Note Regarding Forward-Looking Statements This presentation contains forward-looking statements of NewLink Genetics that involve substantial risks and uncertainties. All statements, other than statements of historical facts, contained in this presentation are forward- looking statements, within the meaning of The Private Securities Litigation Reform Act of 1995. The words "anticipate," "believe," "estimate," "expect," "intend," "may," "plan," "target," "potential," "will," "could," "should," "seek" or the negative of these terms or other similar expressions are intended to identify forward-looking statements, although not all forward-looking statements contain these identifying words. These forward-looking statements include, among others, statements about NewLink Genetics' financial guidance for 2018; results of its clinical trials for product candidates; its timing of release of data from ongoing clinical studies; its plans related to execution of clinical trials; plans related to moving additional indications into clinical development; NewLink Genetics' future financial performance, results of operations, cash position and sufficiency of capital resources to fund its operating requirements; and any other statements other than statements of historical fact. Actual results or events could differ materially from the plans, intentions and expectations disclosed in the forward-looking statements that NewLink Genetics makes due to a number of important factors, including those risks discussed in "Risk Factors" and elsewhere in NewLink Genetics' Annual Report on Form 10-K for the year ended December 31, 2017 and other reports filed with the U.S. Securities and Exchange Commission (SEC). The forward-looking statements in this presentation represent NewLink Genetics' views as of the date of this presentation. NewLink Genetics anticipates that subsequent events and developments will cause its views to change. However, while it may elect to update these forward-looking statements at some point in the future, it specifically disclaims any obligation to do so. You should, therefore, not rely on these forward-looking statements as representing NewLink Genetics' views as of any date subsequent to the date of this presentation. 2


 
IDO Pathway a Key Immuno-oncology Target 3 Treg, regulatory T cell; IDO, indoleamine 2,3-dioxygenase; MDSC, myeloid-derived suppressor cell; CTL, cytotoxic T lymphocyte. 1. Metz R. Oncoimmunology. 2012;1(9):1460-1468. 2. Johnson TS. Immunol Invest. 2012;41(6-7):765-797.


 
IDO Pathway a Key Immuno-oncology Target 4Treg, regulatory T cell; IDO, indoleamine 2,3-dioxygenase; MDSC, myeloid-derived suppressor cell; CTL, cytotoxic T lymphocyte.


 
IDO Expression in Certain Tumors is Associated with Poor Patient Outcomes 5 IDO, indoleamine 2,3-dioxygenase; LN, lymph node; NSCLC, non-small cell lung cancer; DLBCL, diffuse large B-cell lymphoma; RCC, renal cell carcinoma; TCC, transitional cell carcinoma; TNBC, triple-negative breast cancer. Munn DH, et al. J Clin Invest. 2004;114(2):280-290.


 
Indoximod Differentiated Mechanism of Action 6IDO, indoleamine 2,3-dioxygenase; Treg, T regulatory cell; DC, dendritic cell. 1. Brincks EL, et al. Poster presented at the AACR Annual Meeting. April 14-18, 2018. Abstract 3753.  Orally administered, small-molecule IDO pathway inhibitor that reverses the immunosuppressive effects of low tryptophan and high kynurenine that result from IDO activity  Immunostimulatory effects involving 3 main cell types: CD8+ T cells, T regulatory cells, and dendritic cells1 – Reverses effects of low tryptophan by increasing proliferation of effector T cells – Directly reprograms T regulatory cells to helper T cells – Downregulates IDO expression in dendritic cells  Potential synergy has been shown with checkpoint blockade, chemotherapy, radiation and vaccines


 
Indoximod vs Epacadostat: A Differentiated Mechanism of Action Indoximod Directly Reprograms T Regulatory Cells Helper T Cells 7 - 1 0 1 2 3 0 2 0 4 0 6 0 8 0 1 0 0 0 2 0 4 0 6 0 8 0 1 0 0 L o g [ I n d o x i m o d ] (  M ) % f o x p 3 + a m o n g C D 4 + C D 2 5 + c e ll s % IL -1 7 + a m o n g C D 4 + c e lls - 4 - 3 - 2 - 1 0 0 1 0 2 0 3 0 4 0 5 0 6 0 l g [ E p a c a d o s t a t ] (  M ) % f o x p 3 + a m o n g C D 4 + C D 2 5 + c e ll s + K Y N C o n t r o l N o K Y N C o n t r o l AACR Poster #3753 (Brincks, EL, et al, 2018) Indoximod Epacadostat


 
Designing Multimodal Chemo-radio-immunotherapy 8  Hypothesis – Immune activation (immunotherapy) can allow responsiveness to chemotherapy and radiation in patients who would otherwise be refractory  However, this synergy with chemotherapy/radiation requires targeting the antigen-presenting step and creating a pro-inflammatory (immunogenic) tumor milieu – Essentially, it must break tolerance to the dying/apoptotic tumor cells – This antigen cross-presentation step lies upstream of the conventional T-cell checkpoints


 
Recurrent/Refractory Pediatric Brain Tumors 9 Recurrent/refractory brain tumors represent the greatest single cause of mortality in pediatric cancer • Cannot be cured by current standard treatments (treatment- refractory) • Standard of care is largely palliative PFS, progression-free survival; OS, overall survival; HGG, high grade glioma. Historical controls adapted from: DeWire M, et al. J Neurooncol. 2015;123:85; Cefalo G, et al. Neuro Oncol. 2014;16:748; Muller K, et al. Radiat Oncol. 2014;9:177; Fangusaro JR, et al. J Clin Oncol. 2017;35(suppl): abstract 10543 Historical control data for relapsed brain tumors


 
First-in-children Phase 1 Trial of Indoximod-based Multimodal Chemo-radio-immunotherapy 10 • Multimodal management is a key feature of the regimen • Radiographic evidence of progression (escape lesions) can be managed with continued indoximod and: – Surgical resection (regain local control) – Targeted radiation (regain local control) – Crossover to 2nd-line chemotherapy (cyclophosphamide/etoposide) • Relapsed or refractory primary brain tumor patients • Primary endpoints – Regimen limiting toxicities of indoximod + temozolomide – Objective response rate – Regimen-limiting toxicities of indoximod + radiation – Safety • Key eligibility criteria – 3-21 years of age – Histologically proven initial diagnosis of primary malignant brain tumor, with no known curative treatment options – MRI confirmation of tumor progression MRI, magnetic resonance imaging. Clinicaltrials.gov (NCT02502708).


 
First-in-children Phase 1 Trial of Indoximod-based Multimodal Chemo-radio-immunotherapy 11 Group 1 • Indoximod dose escalation (study dose, PO, twice daily on days 1-28) • Temozolomide (200 mg/m2/day, PO, once daily on days 1-5 of 28-day cycles) Group 2 (expansion cohort of Group 1) • RP2D of indoximod • Temozolomide (200 mg/m2/day, PO, once daily on days 1-5 of 28-day cycles) Group 3 • Indoximod dose escalation (study dose, PO, twice daily on days 1-28) • Individualized radiation plan • Followed by indoximod combined with cyclic temozolomide Group 4 (progressive disease on indoximod + temozolomide) • Indoximod (32 mg/kg/dose PO, twice daily on days 1-28) • Cyclophosphamide (2.5 mg/kg/dose PO, once daily) • Etoposide (50 mg/m2/dose PO, once daily) PO, orally; RP2D, recommended phase 2 dose. Clinicaltrials.gov (NCT02502708


 
Patient Demographics (Mixed Population) 12 *Includes one each gliosarcoma, bithalamic glioma, and ganglioglioma. **Includes one previously classified as primitive neuroectodermal tumor. Total patients enrolled N = 29 Diagnosis, n (%) Ependymoma Malignant glioma* Medulloblastoma** 14 (48) 9 (31) 6 (21) Gender, n (%) Female Male 10 (34) 19 (66) Race, n (%) African American Caucasian Hispanic Other Declined to provide 3 (10) 23 (79) 0 2 (7) 1 (3) Age, years Median Range 12.5 320


 
Patient 001: Example of Multimodal Management Chemo-radio-immunotherapy 13 (TTRF) Continuing indoximod- based therapy Progressing disease Responding/stable disease Chemo regimen 1 Chemo regimen 2 Chemo regimen 3 Surgery PR PD PR PDMRI  Pt 001 SD Surgery Chemo 1 Chemo 2 Chemo 3 0 6 12 18 24 Continuous Indoximod (months) Partial RT (SRS) Low-dose outpatient chemo Partial RT (low-dose) RT RT RT • 7-year-old with ependymoma: prolonged disease responsiveness • Indoximod-based multimodal regimen is well tolerated SRS, stereotactic radiosurgery; RT, radiation therapy; PR, partial response; PD, progressive disease; SD, stable disease.


 
Patient 001: Continued Responsiveness Using Indoximod-based Multimodal Management 14 indoximod + radiation tumor (20 Gy) indoximod + 3rd-line chemo indoximod + 3rd-line chemo NOT TARGETED WITH NEW RADIATION Johnson, T, et al, AACR 2018


 
Radio-Immunotherapy Improves Time to Regimen Failure (TTRF) 15 Historical controls adapted from: DeWire M, et al. J Neurooncol 2015;123:85. RT, radiation therapy Fangusaro JR, et al. J Clin Oncol, 2017;35(suppl): abstract 10543. Cefalo G, et al. Neuro Oncol 2014;16:748. Muller K, et al. Radiat Oncol 2014;9:177.


 
New Metastatic Tumor Arising While on Therapy Later Regresses 16 CSF, cerebrospinal fluid; TTRF, time to regimen failure. Pretreatment 2 cycles 4 cycles 6 cycles Begin indoximod + temozolomide 14 yo with CSF relapse of medulloblastoma Potential for late responses makes TTRF an important outcome metric


 
Indoximod-based Multimodal Regimen is Well Tolerated 17  In the 29 patients included in the study, SAEs possibly related to indoximod included 1 case each of: – Febrile neutropenia – Hemiparesis – Hydrocephalus – Spinal cord compression – Status epilepticus – Urinary tract infection  Overall, indoximod did not worsen the toxicity of the base treatment


 
Pilot Cohort in Diffuse Intrinsic Pontine Glioma (DIPG) 18 Group 1 • Indoximod dose escalation (study dose, PO, twice daily on days 1-28) • Temozolomide (200 mg/m2/day, PO, once daily on days 1-5 of 28-day cycles) Group 2 (expansion cohort of Group 1) • RP2D of indoximod • Temozolomide (200 mg/m2/day, PO, once daily on days 1-5 of 28-day cycles) Group 3 • Indoximod dose escalation (study dose, PO, twice daily on days 1-28) • Individualized radiation plan • Followed by indoximod combined with cyclic temozolomide Group 4 (progressive disease on indoximod + temozolomide) • Indoximod (32 mg/kg/dose PO, twice daily on days 1-28) • Cyclophosphamide (2.5 mg/kg/dose PO, once daily) • Etoposide (50 mg/m2/dose PO, once daily) Pilot cohort • Patients with radiographic diagnosis or histologically proven DIPG


 
DIPG Is Rapidly Fatal 19  DIPG has the worst prognosis of any pediatric cancer Median time to progression after radiation is ~6 months1  At progression, patients follow a rapidly declining course – Median OS is 10-12 months2 –Uniformly fatal DIPG, diffuse intrinsic pontine glioma; OS, overall survival. 1. Wolff JE, et al. J Neurooncol. 2012;106(2):391-397. 2. Cohen KJ, et al. Neuro Oncol. 2011;13(4):410-416.


 
Effective Treatments for DIPG are Lacking 20  Standard-of-care treatment is palliative radiation (usually 54 Gy) Chemotherapy has no proven benefit  Thus far, trials have not shown clinical benefit from currently available chemotherapy, radiosensitizing drugs, or biologics  Due to their location in the brainstem, DIPGs cannot be surgically removed DIPG, diffuse intrinsic pontine glioma.


 
Multimodal Chemo-radio-immunotherapy for DIPG Pilot Cohort 21  First question: Could DIPG patients tolerate the indoximod immunotherapy regimen? – DIPG patients are often highly symptomatic  Pilot cohort of 6 newly diagnosed DIPG patients – All 6 patients have finished upfront radiation combined with indoximod – All 6 patients showed initial improvement in symptoms – 3/6 later developed inflammatory symptoms (eg, waxing/waning, migratory) • 2 of these occurred during first cycle of temozolomide with indoximod .


 
NLG2105-037: 9.4-Year-Old Male with Newly Diagnosed DIPG 22 . Serial sections on MRI (T2 Flair)Baseline (pretreatment) After 6 weeks of indoximod + radiation (54 Gy) This patient is neurologically normal at 6 months DIPG scans reviewed by Tina Young-Poussaint, M.D., Boston Children’s Hospital Patient 037 classified as: “Significant response” DIPG, diffuse intrinsic pontine glioma; MRI, magnetic resonance imaging..


 
NLG2105-035: 9.3-Year-Old Male with Newly Diagnosed DIPG 23 . DIPG, diffuse intrinsic pontine glioma; MRI, magnetic resonance imaging.. This patient has sustained neurological improvement at 6 months Serial sections on MRI (T2 Flair) Baseline (pretreatment) After 6 weeks of indoximod + radiation (54 Gy)


 
Additional Newly Diagnosed DIPG Patients 24 . DIPG, diffuse intrinsic pontine glioma This patient has sustained neurological improvement at 6 months Serial sections on MRI (T2 Flair) Baseline (pretreatment) After 6 weeks of indoximod + radiation (54 Gy)


 
Additional Newly Diagnosed DIPG Patients 25 . DIPG, diffuse intrinsic pontine glioma Baseline (pretreatment) After 6 weeks of indoximod + radiation (54 Gy) NLG2105-042 12 yo male NLG2105-043 15 yo female NLG2105-047 5 yo female NLG2105-048 6 yo female


 
Conclusions and Future Directions 26  Phase 1 data suggest that indoximod-based immunotherapy can allow disease responsiveness to conventional therapy (radiation, chemotherapy)  Pilot cohort is under way applying this approach to newly diagnosed DIPG patients  Phase 2 trial is planned .