46
Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences Clinical Research Program Leader, Gastrointestinal Cancers Program Yale Cancer Center New Haven, Connecticut

Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

Embed Size (px)

Citation preview

Page 1: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

Key Concepts in Targeting Cancer Stem Cells

to Manage Disease

Key Concepts in Targeting Cancer Stem Cells

to Manage Disease

ModeratorHoward S. Hochster, MD Professor of MedicineAssociate Director for Clinical SciencesClinical Research Program Leader, Gastrointestinal Cancers ProgramYale Cancer CenterNew Haven, Connecticut

Page 2: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

Max S. Wicha, MDMadeline and Sidney Forbes Professor of OncologyFounding Director EmeritusUniversity of Michigan Comprehensive Cancer CenterAnn Arbor, Michigan

Manish A. Shah, MD Associate Professor of Gastrointestinal OncologyDirector, Gastrointestinal Oncology ProgramCo-Director, Center for Advanced Digestive CareWeill Cornell Medical College of Cornell University/New York-Presbyterian HospitalNew York, New York

Panelists

Page 3: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

Program GoalsProgram Goals

• Define cancer stem cells (CSCs)

• Understand the role of CSCs in tumor development, metastasis, and disease progression

• Describe how CSCs resist chemotherapy and other conventional treatments

• Review advances in CSC-directed therapies

This program will discuss investigational agents that the FDA has not approved for use in the United States.

Page 4: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

What Are CSCs?What Are CSCs?

• CSCs (sometimes called tumor-initiating cells) are a small subpopulation of cells in a tumorb

• CSCs were first identified in AML and have since been found in most solid tumors and hematologic cancersb,c

• According to the CSC hypothesis, CSCs drive tumorigenesis and metastasisa

a. Clarke MF, et al. Cancer Res. 2006;66:9339-9344; b. Bonnet D, Dick JE. Nat Med. 1997;3:730-737; c. Al-Haji M, et al. Proc Natl Acad Sci U S A. 2003;100:3983-3988.

"CSCs are cells within a tumor that possess the capacity to self-renew and to cause the heterogeneous lineages of cancer cells that comprise the tumor."

- AACR Consensus Workshop, 2006a

Page 5: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

Types of Human Stem CellsTypes of Human Stem Cells

• Pluripotent stem cells that can give rise to any type of cell in the body

• Found in embryos• Capable of self-renewal

Embryonic stem cells

• Multipotent stem cells that can differentiate into all the specialized cells needed to maintain their dedicated tissue

• Found in all organs and in the blood• Capable of self-renewal

Adult stem cells

• Multipotent stem cells that can differentiate into all the cells within a tumor

• Found in the tumor microenvironment or "niche"

• May arise from a mutated ASC or progenitor cell or from a reprogrammed differentiated cell

Cancer stem cells

De Los Angeles A, et al. Nature. 2015;525:469-478.

Page 6: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

• This is the gold standard for definitively identifying CSCsa

• Suspected CSCs are injected into immunodeficient mice to see whether they generate tumorsa

− Only a few CSCs are needed to initiate a tumor − Drawback is differences in mouse vs human immune system

Detecting CSCsDetecting CSCs

Cell Surface Markers• Fluorescence-activated cell sorting and conjugating antibodies to

magnetic beads can identify CSC markers• Colony-forming assays show whether a cell can self-renew• Sphere-forming assays measure how many tumor spheres

a CSC produces in vitro (tumorigenic efficiency)b

• Cell surface markers are good surrogates but are not definitive a. Bonnet D, Dick JE. Nat Med. 1997;3:730-737; b. Ajani JA, et al. Semin Oncol. 2015;42:S3-S17.

Serial Transplantation Assays

Page 7: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

CSC Markers in Different Tumor TypesCSC Markers in Different Tumor Types

a. Ajani JA, et al. Semin Oncol. 2015;42:S3-S17; b. Grosse-Gehling P, et al. J Pathol. 2013;229:355-378; c. De Beca FF, et al. J Clin Pathol. 2013;66:187-191.

• CD133+ is 1 of the most common CSC markersa,b

− CD133+ CSCs were isolated first in brain tumors and later in cancers of the colon, liver, breast, lung, ovary, prostate, pancreas, eye, skin, and bone/soft tissue

− CD133+ may predict chemoresistance and worse survival

• Breast CSC markers include CD44+, CD24-, and ALDH− Markers vary depending on histologic subtypec

− CD44+/CD24-/low phenotype predicts poor prognosis

• Some markers (eg, CD44 and CD24) are observed in CSCs across many tumor typesa

Page 8: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

CSC Model vs Stochastic ModelCSC Model vs Stochastic Model

CSC

CSC

CSC CSC

CSC

CSC

CSC

CSC

CSC

CSC

CSC

P

P

P P

P P

P

M M

M M M M M M

Asymmetricdivision

Symmetricdivision

Stochastic (Clonal Evolution) Model

= oncogenic event

CSC (Hierarchical) Model

= oncogenic eventCSC = cancer stem cellP = progenitor cellM = mature cell

All tumor cells can self-renew or differentiate.

Page 9: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

Tumor HeterogeneityTumor Heterogeneity

Shackleton M, et al. Cell. 2009;138:822-829.

• Both the stochastic and CSC models start with 1 cell that produces a tumor with genetically distinct cells• The stochastic model attributes genetic heterogeneity in

tumors to clonal evolution, with no cell hierarchy • The CSC model also accounts for tumor heterogeneity− Mutations or epigenetic changes cause CSC subclones to

produce different progenitor cells from their parents− Mature cells with acquired mutations dedifferentiate into stem-

like cells and proliferate− Migration of a CSC into another tissue microenvironment can

cause further diversification

Page 10: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

Seed-and-Soil HypothesisSeed-and-Soil Hypothesis

a. Paget S. Cancer Metastasis Rev. 1989;8:98-101; b. Langley RR, Fidler IJ. Int J Cancer. 2011;128:2527-2535; c. Chambers AF, et al. Nat Rev Cancer. 2002;2:563-572; d. Folkman J. N Engl J Med. 1971;285;1182-1186.

• In 1889, Paget noticed metastasis was more frequent in certain organs and proposed it was not randoma-c

• Studies suggest metastasis results from signaling between the cancer cell (seed) and the organ microenvironment at metastatic sites (soil)b

• The microenvironment regulates angiogenesis, an important component of metastasisd

"When a plant goes to seed, its seeds are carried in all directions; but they can only live and grow if they fall on congenial soil."

– Stephen Paget, 1889a,b

Page 11: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

Epithelial-to-Mesenchymal Transition (EMT)Epithelial-to-Mesenchymal Transition (EMT)

a. Radisky DC, LaBarge MA. Cell Stem Cell. 2008;2:511-512; b. Thiery JP. Nat Rev Cancer. 2002;2:442-454.

• EMT is important during embryogenesis and wound repaira

• Loss of epithelial markers like E-cadherin (CDH1) causes CSCs to undergo EMT and lose their adhesive propertiesb

− The cells detach from the extracellular matrix and from each othera,b

− They progressively acquire mesenchymal-like traitsa,b

− The now free-floating mesenchymal cells enter the blood stream or lymph nodes and metastasize to distant sitesa

Epithelial Cells

Transition

Mesenchymal Cells

Basement membrane

Page 12: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

Plasticity of CSCs and EMTPlasticity of CSCs and EMT

Liu S, et al. Stem Cell Reports. 2013;2:78-91.

The plasticity of CSCs allows the cells to transition between epithelial and mesenchymal states, which is what gives them the ability to invade tissue, disseminate, and grow at metastatic sites.

Epithelial-like State Mesenchymal-like State• Express epithelial markers• Have polarity• Proliferate extensively• Found more in tumor interior

• Express mesenchymal markers• Remain relatively quiescent• Found at tumor invasive front• Invade the bloodstream• Establish micrometastases

Qualities of CSCs in Epithelial-like vs Mesenchymal-like State

Page 13: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

Drivers of Epithelial-Mesenchymal and Mesenchymal-Epithelial TransitionsDrivers of Epithelial-Mesenchymal and Mesenchymal-Epithelial Transitions

a. Liu S, et al. Stem Cell Reports. 2013;2:78-91; b. Thiery JP. Nat Rev Cancer. 2002;2:442-454; c. Kaplan RN, et al. Nature. 2005;438:820-827.

• Reversible EMT/MET transitions are regulated by the tumor microenvironment• Adverse tumor conditions that drive EMT includea,b

− Hypoxia (sometimes caused by antiangiogenic agents)− Nutrient deprivation− Mutations that cause E-cadherin loss

• microRNA regulates EMT/MET via multiple pathwaysb

• Myeloid precursor cells at the pre-metastatic site (niche) send homing signals to attract CSCsc

Page 14: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

Therapeutic Targets for CSCsTherapeutic Targets for CSCs

a. Ajani JA, et al. Semin Oncol. 2015;42:S3-S17; b. Iliopoulous D, et al. Cell. 2009;139:693-706; c. Ginestier C, et al. J Clin Invest. 2010;120:485-497.

• Therapies should target stemness pathways (internal pathways that induce or maintain stemness properties in CSCs)a

− Wnt receptors and ligands− Notch receptors and ligands− Hedgehog (ligands, Patched-Smoothened complex)− JAK/STAT pathway, including STAT3− TGF-β family of cytokines− Hippo− Crosstalk between Notch and other oncogenic pathways

• Inflammatory mediators (extracellular pathways), like IL-6 and IL-8, promote CSC self-renewal and are also good treatment targetsb,c

Page 15: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

Response of CSCs to ChemotherapyResponse of CSCs to Chemotherapy

a. Ginestier C, et al. J Clin Invest. 2010;120:485-487; b. Li X, et al. J Natl Cancer Inst. 2008;100:672-679.

• In preclinical models, the CSC population increased after chemotherapya

• Clinical studies have observed enrichment of CSCs in patients treated with chemotherapyb

• One proposed mechanism is the cytokines that dying cells release (eg, IL8) stimulate development of CSCsa

CSC

Popu

latio

n, %

Control

Repar

ixin

Doceta

xel

Repar

ixin/

Doceta

xel

20

16

12

8

4

0

In Vitro Assessment of CSCs After Chemotherapya

Page 16: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

Pilot Study of Reparixin Plus PaclitaxelPilot Study of Reparixin Plus Paclitaxel

Schott AF, et al. SABCS 2014. Abstract P6-03-01.

• Phase 1b study in HER2-negative MBC (N = 33)− Reparixin loading dose (400, 800, or 1200 mg) 3 times/d for 3

days was followed by paclitaxel (80 mg/m2/wk) + reparixin for 21 days

− Treatment continued until disease progression or toxicity

• Safety was assessed after 1 cycle− 15 serious AEs occurred, none related to reparixin− 30% of patients experienced a grade 3/4 AE

• Efficacy assessment included 18 patients− 2 CR and 6 PR− Duration of response: 2-20 mo

Page 17: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

Co-Targeting CSCs and Bulk Tumor CellsCo-Targeting CSCs and Bulk Tumor Cells

• Targeting CSCs− CSCs that survive treatment will repopulate the tumor− CSCs can cause recurrence even years later− Surviving CSCs have the potential to metastasize

• Targeting bulk non-tumorigenic cells− In advanced cancer, the bulk tumor population is large

enough to constitute a huge burden or death− Plasticity may allow mature tumor cells to

dedifferentiate and acquire stem-like properties

Effective treatment strategies must target the CSCs and the cells that make up the bulk of the tumor.

Page 18: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

Resistance of CSCs to Conventional TherapyResistance of CSCs to Conventional Therapy

a. Maugeri-Saccà M, et al. Clin Cancer Res. 2011;17:4942-4947; b. Skvortsova I, et al. SeminCancer Biol. 2015 Sep 22. [Epub ahead of print]; c. Hashida S, et al. Cancer Sci. 2015 Jul 21. [Epub ahead of print]

CSCs have several pro-survival mechanisms that help them resist chemotherapy, radiotherapy, and targeted drugs.

Intrinsic Mechanisms Indirect Mechanisms• Enhanced DNA repair• High expression of drug

efflux pumps• Upregulation of anti-

apoptotic factors and detoxification enzymes• Quiescence during treatment

to protect self-renewal ability

• Induction of EMT or acquisition of CSC markers via signaling pathways in the microenvironment• Hypoxia-triggered adaptive

changes via HIF activation (eg, formation of new blood vessels that are poor drug transporters)

Page 19: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

Tumor Regression: Inadequate End PointTumor Regression: Inadequate End Point

Reya T, et al. Nature. 2001;414:105-111.

CSC-directed

therapy

Conventional therapy

Tumor cannot make new cells Mature cells die,

tumor shrinks, patient is cured.

CSC regenerates the tumor Patient has a relapse.

CSC

CSC

CSCNotable tumor

shrinkage

Little tumor shrinkage

Tumor shrinkage by RECIST may not be useful for evaluating response to CSC-directed therapies.

Page 20: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

Efficacy End Points for CSC-Directed AgentsEfficacy End Points for CSC-Directed Agents

End PointTime to progression (TTP)

CSC agents delay TTP, so patients will have to take them until they have significant progression to determine their effectiveness

Pathologic CR to neoadjuvant therapy

This is a well-defined predictor of recurrence; a biopsy before and after neoadjuvant treatment would show the direct effects of therapy on CSCs

Immune-related response criteria

Measurable new lesions are incorporated into total tumor burden because tumors may grow before response is evident

Reduction in CSCs A decrease in CSCs may predict better prognosis

Overall survival CSC-directed agents must be shown to prolong survival without compromising patient safety

Possible End Points for Trials of CSC-Directed Agents

Page 21: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

CSCs as a Prognostic MarkerCSCs as a Prognostic Marker

a. Ajani JA, et al. Semin Oncol. 2015;42:S3-S17; b. Charafe-Jauffret E, et al. Clin Cancer Res. 2010;16:45-55; c. Sakakibara M, et al. Cancer. 2012;118:3899-3910; d. Khoury T, et al. Mod Pathol. 2012;25;388-397.

• Studies have linked CSC markers with an increased risk of metastasis or recurrence, worse survival outcomes, and treatment resistancea

• CSC markers have been found to have prognostic significance in many solid tumors, including breast, colon, brain, and head and neck cancers; and in hematologic malignancies such as AMLa

• Breast cancer studies have shown higher proportions of cells expressing CSC marker ALDH1+ after neoadjuvant chemotherapy predicts early metastasis and worse survival outcomesb-d

Page 22: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

Circulating Tumor Cells and CSCsCirculating Tumor Cells and CSCs

a. Balic M, et al. Expert Rev Mol Diagn. 2012;12:303-312; b. Aktas B, et al. Breast Cancer Res. 2009;11:R46; c. Karabacak NM, et al. Nat Protoc. 2014;9:694-710.

• Circulating tumor cells (CTCs) can predict the risk of metastasis and relapse in patients treated for cancera

• Analyzing the portion of CTCs that are CSCs may offer clues to the efficacy of CSC-directed therapies• The only FDA-approved method for isolating CTCs

uses the epithelial adhesion molecule EpCAMa

− ~50% of CTCs have undergone EMT and have low expression of EpCAM or other epithelial markersb

− This method may fail to detect CSCs that survive therapy

• New technologies are being studied that use microfluidics to isolate single cells by biomarkerc

Page 23: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

CSC-Directed Agents in Clinical TrialsCSC-Directed Agents in Clinical Trials

a. Liu S, Wicha MS. J Clin Oncol. 2010;28:4006-4012; b. Prud'homme GJ. Curr Pharm Des. 2012;18:2838-2849.

Tarextumab(OMP-59R5)

β-CAT

IIIIIIII

IIIIIIII

IIIiIIIii

iiiiiiiiii

iiiiiiiiii

iIL-

8II

CXCR1

Self-renewal, drug resistance, metastasisTARGET DNA

Cancer stem cell

NOTCH

DLL/JAG WNT

LPR/FZD

Demcizumab(anti-DLL4)

Vantictumab(anti-FZD)

Ipafricept(OMP-54F28)

Reparixin

FAK Defactinibβ-CAT, STAT3, NanogBBI608

IL-8

CXCR1

FAK

Several CSC-directed agents with various targets were shown to be safe in phase 1 clinical trials.

Page 24: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

Forced Differentiation of CSCsForced Differentiation of CSCs

a. Guessous F, et al. Cell Cycle. 2010;9:1031-1036.

• CSC-directed agents that inhibit signaling along the self-renewal pathway have been found to activate quiescent CSCs, causing them to differentiatea

• Inducing CSCs to differentiate may represent a viable CSC-directed treatment strategy − Once differentiated, the CSCs would be susceptible to

chemotherapy− You could potentially exhaust the population of CSCs

• Effective cancer treatment will likely require a combination regimen with a CSC-directed agent and an agent that targets the bulk non-tumorigenic cells

Page 25: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

Immunotherapy and CSCsImmunotherapy and CSCs

a. Teitz-Tennenbaum S, et al. Oncoimmunology. 2012;1:1401-1403; b. Lin M, et al. Immunol Res. 2015;62:16-22.

• A small percentage of patients with cancer treated with immunotherapy have durable remission• Preclinical models suggest the immune system in

these patients eradicates the CSCs• Anti-CSC vaccines are in development− In a proof-of-concept study, a dendritic cell-based vaccine

gave immunocompromised mice antitumor immunitya

− Preliminary evidence from a phase 1/2 study (N = 90) of a novel lung cancer CSC vaccine found it safe and effectiveb

• Combining vaccines with checkpoint inhibitors may improve cure rates in some cancers

Page 26: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

SummarySummary

• Growing evidence shows CSCs contribute to the development, growth, and metastasis of cancer• CSCs may originate from a mutated ASC or a mature

cell that acquires stem-like properties• Many novel agents that target CSC signaling

pathways or cytokines are being studied in clinical trials, and early data suggest these drugs are relatively safe• More research is needed to identify appropriate end

points for CSC-directed therapies and to identify safe combinations that target CSCs and bulk tumor cells

Page 27: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

AbbreviationsAbbreviationsAE = adverse eventALDH = aldehyde dehydrogenase AML = acute myelogenous leukemia ASC = adult stem cellCDH1 = cadherin-1CR = complete responseCSC = cancer stem cellsCTC = circulating tumor cellEMT = epithelial-to-mesenchymal transitionEpCAM = epithelial cell adhesion moleculeERK= extracellular signal-regulated kinase IL = interleukinGI = gastrointestinal HER2 = human epidermal growth factor receptor 2JAK = Janus kinaseM = mature cellMAPK = mitogen-activated protein kinase

Page 28: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

Abbreviations (cont)Abbreviations (cont)MBC = metastatic breast cancerMSI = microsatellite instabilityP = progenitor cellPD-1 = programmed cell death 1PD-L1 = programmed death ligand 1PR = partial responseRECIST = Response Evaluation Criteria In Solid TumorsSTAT = signal transducer and activator of transcriptionTGF-β = transforming growth factor-βTTP = time to progression

Page 29: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

Abbreviations (cont)Abbreviations (cont)MBC = metastatic breast cancerMSI = microsatellite instabilityP = progenitor cellPD-1 = programmed cell death 1PD-L1 = programmed death ligand 1PR = partial responseRECIST = Response Evaluation Criteria In Solid TumorsSTAT = signal transducer and activator of transcriptionTGF-β = transforming growth factor-βTTP = time to progression

Page 30: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

ReferencesReferences1. Clarke MF, Dick JE, Dirks PB, et al. Cancer stem cells -- perspectives on current

status and future directions: AACR workshop on cancer stem cells. Cancer Res. 2006;66:9339-9344.

2. Odorico JS, Kaufman DS, Thomson JA. Multilineage differentiation from human embryonic stem cell lines. Stem Cells. 2001;19:193-204.

3. Pera MF, Reubinoff B, Trounson A. Human embryonic stem cells. J Cell Sci. 2000;113:5-10.

4. Thompson LH, Björklund A. Reconstruction of brain circuitry by neural transplants generated from pluripotent stem cells. Neurobiol Dis. 2015;79:28-40.

5. Montalbán-Loro R, Domingo-Muelas A, Bizy A, Ferrón SR. Epigenetic regulation of stemness maintenance in the neurogenic niches. World J Stem Cells. 2015;7:700-710.

Page 31: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

References (cont)References (cont)6. Forbes SJ, Gupta S, Dhawan A. Cell therapy for liver disease: From liver

transplantation to cell factory. J Hepatol. 2015;62:S157-S169.

7. Zhou Q, Li L, Zhao B, Guan KL. The hippo pathway in heart development, regeneration, and diseases. Circ Res. 2015;116:1431-1447.

8. Mae S, Osafune K. Kidney regeneration from human induced pluripotent stem cells. Curr Opin Organ Transplant. 2015;20:171-177.

9. Bonnet D, Dick JE. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med. 1997;3:730-737.

10. Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A. 2003;100:3983-3988. Erratum in: Proc Natl Acad Sci U S A. 2003;100:6890.

Page 32: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

References (cont)References (cont)11. Ginestier C, Hur MH, Charafe-Jauffret E, et al. ALDH1 is a marker of normal

and malignant human mammary stem cells and a predictor of poor clinical outcome. Cell Stem Cell. 2007;1:555-567.

12. Houghton J, Stoicov C, Nomura S, et al. Gastric cancer originating from bone marrow-derived cells. Science. 2004;306:1568-1571.

13. Chaffer CL, Brueckmann I, Scheel C, et al. Normal and neoplastic nonstem cells can spontaneously convert to a stem-like state. Proc Natl Acad Sci U S A. 2011;108:7950-7955.

14. Shackleton M, Quintana E, Fearon ER, Morrison SJ. Heterogeneity in cancer: cancer stem cells versus clonal evolution. Cell. 2009;138:822-829.

15. Kaplan RN, Riba RD, Zacharoulis S, et al. VEGFR1-positive haematopoietic bone marrow progenitors initiate the premetastatic niche. Nature. 2005;438:820-827.

Page 33: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

References (cont)References (cont)16. Langley RR, Fidler IJ. The seed and soil hypothesis revisited -- the role of

tumor-stroma interactions in metastasis to different organs. Int J Cancer. 2011;128:2527-2535.

17. Paget S. The distribution of secondary growths in cancer of the breast. 1889. Cancer Metastasis Rev.1989;8:98-101.

18. Chambers AF, Groom AC, MacDonald IC. Dissemination and growth of cancer cells in metastatic sites. Nat Rev Cancer. 2002;2:563-572.

19. Folkman J. Tumor angiogenesis: therapeutic implications. N Engl J Med. 1971;285:1182-1186.

20. Ribatti D. Judah Folkman, a pioneer in the study of angiogenesis. Angiogenesis. 2008;11:3-10.

21. Thiery JP. Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer. 2002;2:442-454.

Page 34: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

References (cont)References (cont)22. Radisky DC, LaBarge MA. Epithelial-mesenchymal transition and the stem

cell phenotype. Cell Stem Cell. 2008;2:511-512.

23. Organ SL, Tsao MS. An overview of the c-MET signaling pathway. Ther Adv Med Oncol. 2011;3: S7-S19.

24. Liu S, Cong Y, Wang D, et al. Breast cancer stem cells transition between epithelial and mesenchymal states reflective of their normal counterparts. Stem Cell Reports. 2013;2:78-91.

25. Thiery JP. Epithelial-mesenchymal transitions in development and pathologies. Curr Opin Cell Bio. 2003;15:740-746.

26. May CD, Sphyris N, Evans KW, Werden SJ, Guo W, Mani SA. Epithelial-mesenchymal transition and cancer stem cells: a dangerously dynamic duo in breast cancer progression. Breast Cancer Res. 2011;13:202.

Page 35: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

References (cont)References (cont)27. Gammon L, Mackenzie IC. Roles of hypoxia, stem cells and epithelial-

mesenchymal transition in the spread and treatment resistance of head and neck cancer. J Oral Pathol Med. 2015 May 7. [Epub ahead of print]

28. Iliopoulos D, Hirsch HA, Struhl K. An epigenetic switch involving NF-kappaB, Lin28, Let-7 MicroRNA, and IL6 links inflammation to cell transformation. Cell. 2009;139:693-706.

29. Ginestier C, Liu S, Diebel ME, et al. CXCR1 blockade selectively targets human breast cancer stem cells in vitro and in xenografts. J Clin Invest. 2010;120:485-497.

30. Li X, Lewis MT, Huang J, et al. Intrinsic resistance of tumorigenic breast cancer cells to chemotherapy. J Natl Cancer Inst. 2008;100:672-679.

31. Tajima H, Ohta T, Kitagawa H, et al. Neoadjuvant chemotherapy with gemcitabine for pancreatic cancer increases in situ expression of the apoptosis marker M30 and stem cell marker CD44. Oncol Lett. 2012;3:1186-1190.

Page 36: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

References (cont)References (cont)32. Mizukami T, Kamachi H, Mitsuhashi T, et al. Immunohistochemical analysis

of cancer stem cell markers in pancreatic adenocarcinoma patients after neoadjuvant chemoradiotherapy. BMC Cancer. 2014;14:687.

33. Barr MP, Gray SG, Hoffmann AC, et al. Generation and characterisation of cisplatin-resistant non-small cell lung cancer cell lines displaying a stem-like signature. PLoS One. 2013;8:e54193.

34. Schott AF, Wicha MS, Perez RP, et al. A phase Ib study of the CXCR1/2 inhibitor reparixin in combination with weekly paclitaxel in metastatic HER2 negative breast cancer -- first analysis. Presented at: 37th Annual CTRC-AACR San Antonio Breast Cancer Symposium; December 9-13, 2014; San Antonio, Texas. Abstract P6-03-01.

35. ClinicalTrials.gov. A Double-Blind Study of Paclitaxel in Combination With Reparixin or Placebo for Metastatic Triple-Negative Breast Cancer (FRIDA). NCT02370238. https://clinicaltrials.gov/ct2/show/NCT02370238. Accessed October 23, 2015.

Page 37: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

References (cont)References (cont)36. Morel AP, Lièvre M, Thomas C, Hinkal G, Ansieau S, Puisieux A. Generation

of breast cancer stem cells through epithelial-mesenchymal transition. PLoS One. 2008;3:e2888.

37. Chaffer CL, Marjanovic ND, Lee T, et al. Poised chromatin at the ZEB1 promoter enables breast cancer cell plasticity and enhances tumorigenicity. Cell. 2013;154:61-74.

38. Skvortsova I, Debbage P, Kumar V, Skvortsov S. Radiation resistance: cancer stem cells (CSCs) and their enigmatic pro-survival signaling. Semin Cancer Biol. 2015 Sep 22. [Epub ahead of print]

39. Maugeri-Saccà M, Vigneri P, De Maria R. Cancer stem cells and chemosensitivity. Clin Cancer Res. 2011;17:4942-4947.

40. Jokinen E, Laurila N, Koivunen P, Koivunen JP. Combining targeted drugs to overcome and prevent resistance of solid cancers with some stem-like cell features. Oncotarget. 2014;5:9295-9307.

Page 38: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

References (cont)References (cont)41. Hashida S, Yamamoto H, Shien K, et al. Acquisition of cancer stem cell-like

properties in non-small cell lung cancer with acquired resistance to afatinib. Cancer Sci. 2015 Jul 21. [Epub ahead of print]

42. Zhou T, Zheng L, Hu Z, et al. The effectiveness of RECIST on survival in patients with NSCLC receiving chemotherapy with or without target agents as first-line treatment. Sci Rep. 2015;5:7683.

43. Edeline J, Boucher E, Rolland Y, et al. Comparison of tumor response by Response Evaluation Criteria in Solid Tumors (RECIST) and modified RECIST in patients treated with sorafenib for hepatocellular carcinoma. Cancer. 2012;118:147-156.

44. Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer, and cancer stem cells. Nature. 2001;414:105-111.

45. Wolchok JD, Hoos A, O'Day S, et al. Guidelines for the evaluation of immune therapy activity in solid tumors: immune-related response criteria. Clin Cancer Res. 2009;15:7412-7420.

Page 39: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

References (cont)References (cont)46. Cortazar P, Zhang L, Untch M, et al. Pathological complete response and

long-term clinical benefit in breast cancer: the CTNeoBC pooled analysis. Lancet. 2014;384:164-172.

47. Charafe-Jauffret E, Ginestier C, Iovino F, et al. Aldehyde dehydrogenase 1-positive cancer stem cells mediate metastasis and poor clinical outcome in inflammatory breast cancer. Clin Cancer Res. 2010;16:45-55.

48. Sakakibara M, Fujimori T, Miyoshi T, et al. Aldehyde dehydrogenase 1-positive cells in axillary lymph node metastases after chemotherapy as a prognostic factor in patients with lymph node-positive breast cancer. Cancer. 2012;118:3899-3910.

49. Alamgeer M, Ganju V, Kumar B, et al. Changes in aldehyde dehydrogenase-1 expression during neoadjuvant chemotherapy predict outcome in locally advanced breast cancer. Breast Cancer Res. 2014;16:R44.

50. Khoury T, Ademuyiwa FO, Chandrasekhar R, et al. Aldehyde dehydrogenase 1A1 expression in breast cancer is associated with stage, triple negativity, and outcome to neoadjuvant chemotherapy. Mod Pathol. 2012;25:388-397.

Page 40: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

References (cont)References (cont)51. Balic M, Lin H, Williams A, Datar RH, Cote RJ. Progress in circulating tumor

cell capture and analysis: implications for cancer management. Expert Rev Mol Diagn. 2012;12:303-312.

52. Gorges TM, Tinhofer I, Drosch M, et al. Circulating tumour cells escape from EpCAM-based detection due to epithelial-to-mesenchymal transition. BMC Cancer. 2012;12:178.

53. Aktas B, Tewes M, Fehm T, Hauch S, Kimmig R, Kasimir-Bauer S. Stem cell and epithelial-mesenchymal transition markers are frequently overexpressed in circulating tumor cells of metastatic breast cancer patients. Breast Cancer Res. 2009;11:R46.

54. Karabacak NM, Spuhler PS, Fachin F, et al. Microfluidic, marker-free isolation of circulating tumor cells from blood samples. Nat Protoc. 2014;9:694-710.

55. Liu S, Wicha MS. Targeting breast cancer stem cells. J Clin Oncol. 2010;28:4006-4012.

Page 41: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

References (cont)References (cont)56. Prud'homme GJ. Cancer stem cells and novel targets for antitumor

strategies. Curr Pharm Des. 2012;18:2838-2849.

57. Pietanza MC, Spira AI, Jotte RM, et al. Final results of phase Ib of tarextumab (TRXT, OMP59R5, antiNotch2/3) in combination with etoposide and platinum (EP) in patients (pts) with untreated extensive stage small-cell lung cancer (ED-SCLC). J Clin Oncol. 2015;33. Abstract 7508.

58. Kotasek D, Hughes BGM, Markman B, et al. A phase 1b study of the anticancer stem cell agent demcizumab (DEM), pemetrexed (PEM) & carboplatin (CARBO) in pts with 1st line non-squamous NSCLC. J Clin Oncol. 2015;33. Abstract 8045.

59. Jimeno A, Gordon MS, Chugh R, et al. A first-in human phase 1 study of anticancer stem cell agent OMP54F28 (FZD8-Fc), decoy receptor for WNT ligands, in patients with advanced solid tumors. J Clin Oncol. 2014;32. Abstract 2505.

Page 42: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

References (cont)References (cont)60. Hitron M, Stephenson J, Chi KN, et al. A phase 1b study of the cancer stem

cell inhibitor BBI608 administered with paclitaxel in patients with advanced malignancies. .J Clin Oncol. 2014;32. Abstract 2530.

61. Jonker DJ, Stephenson J, Edenfield WJ, et al. A phase I extension study of BBI608, a first-in-class cancer stem cell (CSC) inhibitor, in patients with advanced solid tumors. J Clin Oncol. 2014;32. Abstract 2546.

62. Becerra C, Stephenson J, Jonker DJ, et al. J Clin Oncol. 2015;33. Abstract 4069.

63. Laurie SA, Jonker DJ, Edenfield WJ, et al. A phase 1 dose-escalation study of BBI503, a first-in-class cancer stemness kinase inhibitor in adult patients with advanced solid tumors. J Clin Oncol. 2014;32. Abstract 2527.

64. Jonker DJ, Laurie SA, Cote GM, et al. Phase 1 extension study of BBI503, a first-in-class cancer stemness kinase inhibitor, in patients with advanced colorectal cancer. J Clin Oncol. 2015;33. Abstract 3615.

Page 43: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

References (cont)References (cont)65. Shah MA, Muro K, Shitara K, et al. The BRIGHTER trial: A phase III

randomized double-blind study of BBI608 + weekly paclitaxel versus placebo (PBO) + weekly paclitaxel in patients (pts) with pretreated advanced gastric and gastroesophageal junction (GEJ) adenocarcinoma. J Clin Oncol. 2015;33. Abstract TPS4139.

66. Hao J, Li TG, Qi X, Zhao DF, Zhao GQ. WNT/beta-catenin pathway up-regulates Stat3 and converges on LIF to prevent differentiation of mouse embryonic stem cells. Dev Biol. 2006;290:81-91.

67. Guessous F, Zhang Y, Kofman A, et al. microRNA-34a is tumor suppressive in brain tumors and glioma stem cells. Cell Cycle. 2010;9:1031-1036.

68. Korkaya H, Liu S, Wicha MS. Regulation of cancer stem cells by cytokine networks: attacking cancer's inflammatory roots. Clin Cancer Res. 2011;17:6125-6129.

69. Ithimakin S, Day KC, Malik F, et al. HER2 drives luminal breast cancer stem cells in the absence of HER2 amplification: implications for efficacy of adjuvant trastuzumab. Cancer Res. 2013;73:1635-1646.

Page 44: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

References (cont)References (cont)70. Hassan KA, Wang L, Korkaya H, et al. Notch pathway activity identifies cells

with cancer stem cell-like properties and correlates with worse survival in lung adenocarcinoma. Clin Cancer Res. 2013;19:1972-1980.

71. Schott AF, Landis MD, Dontu G, et al. Preclinical and clinical studies of gamma secretase inhibitors with docetaxel on human breast tumors. Clin Cancer Res. 2013;19:1512-1524.

72. Lee Y, Sunwoo J. PD-L1 is preferentially expressed on CD44+ tumor-initiating cells in head and neck squamous cell carcinoma. Poster presented at: Society for Immunotherapy of Cancer 29th Annual Meeting; November 6-9, 2014; National Harbor, Maryland. J ImmunoTherapy Cancer 2014, 2(Suppl 3):P270.

73. Zhi Y, Mou Z, Chen J, et al. B7H1 expression and epithelial-to-mesenchymal transition phenotypes on colorectal cancer stem-like cells. PLoS One. 2015;10:e0135528.

74. Yang Y, Wu KE, Zhao E, et al. B7-H1 enhances proliferation ability of gastric cancer stem-like cells as a receptor. Oncol Lett. 2015;9:1833-1838.

Page 45: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

References (cont)References (cont)75. Teitz-Tennenbaum S, Wicha MS, Chang AE, Li Q. Targeting cancer stem cells

via dendritic-cell vaccination. Oncoimmunology. 2012;1:1401-1403.

76. Deonarain MP, Kousparou CA, Epenetos AA. Antibodies targeting cancer stem cells: a new paradigm in immunotherapy? MAbs. 2009;1:12-25.

77. Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366:2443-2454.

78. Di Giacomo AM, Calabrò L, Danielli R, et al. Long-term survival and immunological parameters in metastatic melanoma patients who responded to ipilimumab 10 mg/kg within an expanded access programme. Cancer Immunol Immunother. 2013;62:1021-1028.

79. Lin M, Li SY, Xu KC, et al. Safety and efficacy study of lung cancer stem cell vaccine. Immunol Res. 2015;62:16-22.

Page 46: Key Concepts in Targeting Cancer Stem Cells to Manage Disease Moderator Howard S. Hochster, MD Professor of Medicine Associate Director for Clinical Sciences

References (cont)References (cont)80. O'Shea JJ, Gadina M, Schreiber RD. Cytokine signaling in 2002: new surprises

in the Jak/Stat pathway. Cell. 2002;109;S121-S131.

81. Parampalli Yajnanarayana S, Stübig T, Cornez I, et al. JAK1/2 inhibition impairs T cell function in vitro and in patients with myeloproliferative neoplasms. Br J Haematol. 2015;169:824-833.

82. Llosa NJ, Cruise M, Tam A, et al. The vigorous immune microenvironment of microsatellite instable colon cancer is balanced by multiple counter-inhibitory checkpoints. Cancer Discov. 2015;5:43-51.