Transcript Document

Current Approaches to the Diagnosis and Management of Non-CML Myeloproliferative Disorders

CME Enduring Material

Current Approaches to the Diagnosis and Management of Non-CML Myeloproliferative Disorders

Review Date:

April, 2003

Release Date:

June, 2003

Credit Available Through:

June, 2005 A Continuing Medical Education Activity

Sponsored by:

Mount Sinai School of Medicine

Supported by:

An unrestricted educational grant from Shire US Inc.

Credit issued by:

Mount Sinai School of Medicine

Produced by:

Jonathan Wood & Associates

This activity was planned and produced in accordance with the ACCME Essentials

Overall Purpose

This educational slide program, with accompanying notes pages and CD ROM, is a compilation of experts’ slides from a series of conferences held around the country on the non-CML myeloproliferative disorders (MPDs). The program is intended to provide the latest findings and a comprehensive background on this rare group of disorders and should serve as a ready-reference for physicians in their own practices.

Target Audience

Hematologists, oncologists, and allied health professionals who care for MPD patients

Objectives

Upon completion of this course, hematologists, oncologists, and allied health professionals should be able to: • Recognize MPDs as stem cell disease • Differentiate between clonal and nonclonal thombocytosis • Describe the pathophysiology of myeloproliferative platelets • Understand leukemic transformation as a natural progression of MPDs or as a sequela of therapy • Evaluate the use of allogeneic and autologous stem cell transplantation for MPDs, including selection of candidates, use of conditioning regimens, and complications of transplantation • Diagnose accurately and treat patients with MPDs in their practice • Choose among the various treatment options • Recognize disturbances of normal hemostatic mechanisms in MPDs

Accreditation

This activity has been planned and implemented in accordance with the Essentials and Standards of the Accreditation Council for Continuing Medical Education (ACCME) through the sponsorship of Mount Sinai School of Medicine. Mount Sinai School of Medicine is accredited by ACCME to provide continuing medical education for physicians. Mount Sinai School of Medicine designates this continuing medical education activity for a maximum of 5 credits in Category 1 credit towards the AMA Physician’s Recognition Award. Each physician should claim only those hours that he/she spent in the educational activity.

Grantor

This activity is made possible by an unrestricted educational grant from Shire US Inc.

Produced By

Jonathan Wood & Associates

Faculty

Steven M. Fruchtman, MD, Chair

Clinical Director, Division of Hematology Mount Sinai School of Medicine New York, New York Disclosure: Grant/Research Support: Merck and Co, Inc, Fujisawa Healthcare Inc, Shire US Inc.; Consultant/Advisory Board: Sangstat Medical Corp, Novartis; Speakers Bureau: Shire US Inc., Merck and Co, Inc

Charles L. Bennett, MD, PhD

Professor of Medicine Northwestern University School of Medicine Chicago, Illinois Disclosure: Consultant/Advisory Board/Speakers Bureau: Shire US Inc.

Harriet S. Gilbert, MD

Clinical Professor of Medicine Albert Einstein College of Medicine Bronx, New York Disclosure: Speakers Bureau: Shire US Inc.

Craig Kessler, MD

Professor of Medicine and Pathology Chief, Division of Hematology-Oncology Georgetown University Medical Center Washington, DC Disclosure: No commercial relationships to disclose

John M. McCarty, MD

Associate Professor Director, Bone Marrow Transplantation Program Medical College of Virginia, Virginia Commonwealth University Richmond, Virginia Disclosure: No commercial relationships to disclose

Robert M. Petitt, MD

Senior Consultant in Hematology Mayo Clinic Rochester, Minnesota Disclosure: Consultant/Advisory Board: Shire UK; Speakers Bureau: Shire US Inc.

General Disclosure

Four current therapeutic alternatives that are being used in the management of myeloproliferative disorders are cited throughout this educational slide program. Anagrelide is the only compound that the FDA has approved for the treatment of patients with thrombocythemia secondary to myeloproliferative disorders. Use of hydroxyurea, interferon  , or stem cell transplant are unproven therapies that do not have an FDA indication in the treatment of the non-CML myeloproliferative disorders.

It is the policy of the Mount Sinai School of Medicine to ensure fair balance, independence, objectivity, and scientific rigor in all its sponsored programs. All faculty participating in sponsored programs are expected to disclose to the audience any real or apparent conflict of-interest related to the content of their presentation, and any discussions of unlabeled or investigational use of any commercial product or device not yet approved in the United States.

Table of Contents Table of Contents

Overview: Epidemiology, Etiology, and Pathophysiology Natural History Platelet Physiology Diagnosis and Diagnostic Issues Treatment

Essential Thrombocythemia

• • •

Polycythemia Vera Idiopathic Myelofibrosis Stem Cell Transplant Cost-effectiveness Conclusions References/Posttest/Evaluation

Overview: Epidemiology, Etiology, and Pathophysiology

Chronic Myeloproliferative Disorders

Four well-characterized subgroups: • Polycythemia vera (P Vera) • Essential thrombocythemia (ET) • Agnogenic myeloid metaplasia (AMM) • Chronic myelogenous leukemia (CML) HS Gilbert

Potentially Confusing Disorders

• P Vera, or CML, or AMM • Undifferentiated chronic myeloproliferative disorder • Cellular agnogenic myeloid metaplasia • Chronic myelomonocytic leukemia • Chronic basophilic, eosinophilic, or neutrophilic leukemia • Juvenile chronic myelogenous leukemia RM Petitt

MyM WBC RBC PLTS

Non-CML MPDs

Pluripotential Hematopoietic Stem Cell Monoclonal Expansion Retention of Pluripotentiality, Commitment, Differentiation, Maturation Over-Production of Functioning Circulating Hematic Populations Erythroid POLYCYTHEMIA VERA THROMBOSIS HEMORRHAGE Megakaryocyte/ Platelets ESSENTIAL THROMBOCYTHEMIA THROMBOSIS HEMORRHAGE Myeloid/Monocyte Mast cell/Basophil "Ph- CML" "CMML-OID" HYPERLEUKOCYTOSIS HYPERHISTAMINEMIA LYSOZYMURIA Extramedullary Hematopoiesis MYELOID METAPLASIA

HS Gilbert

MyM WBC RBC PLTS

Phenotypes of MPDs

ET P Vera MyM

HS Gilbert

Causes of High Platelet Counts

• Essential thrombocythemia • Other chronic myeloproliferative disorders – P Vera, MM-MF, CML • Reactive thrombocytosis • Familial thrombocythemia – High TPO, normal TPO (?abnormal c-mpl) • Myelodysplastic syndromes – CMML, 5q minus • Laboratory artifact – ANLL RM Petitt

ET: Pathophysiology

• Platelet production increased more than 5-fold – 237 million (normal 43 million) per day • Platelet survival normal • Megakaryocyte mass increased almost 4-fold – Number increased – Volume increased – Diameter increased (reflects increased ploidy) • Megakaryocyte ploidy increased – 64N and 128N cells relatively common RM Petitt

Reactive Thrombocytosis

• Acute or chronic inflammatory disease • Acute or chronic bleeding • Iron deficiency • Chronic marrow stimulation, e.g. hemolysis • Post-thrombocytopenic rebound • Disseminated malignancy • Absence of spleen (surgical, congenital, functional) • Intense exertion, parturition, trauma, epinephrine RM Petitt

Clinical Features of ET

• Median age at diagnosis = 60 years • Between 25 and 50 years, more common in women; young women in childbearing age are a subgroup with special problems • Microvascular symptoms are common; thrombotic complications exceed bleeding complications • < 5% incidence of transformation into myelofibrosis with myeloid metaplasia or acute leukemia HS Gilbert

Thrombosis in ET

• Thrombotic risk per Cortelazzo et al: – 30% in patients with prior history of thrombosis – 3% in patients with no prior history of thrombosis • Thrombotic complications in one cohort was 6.6 per patient-year (higher in > 60 y/o and those with prior history of thrombosis) HS Gilbert

Thrombosis in ET

• Increased incidence of thrombotic complications in treated patients with inadequately controlled platelet counts – Current data suggest that thrombotic complications can and do occur at relatively low platelet levels and recommend reduction of platelets to <400,000/ m L HS Gilbert

Incidence of ET

120 100 80 60 40 20 Men Women 0 <10 10 –15 16–20 21–25 26–30 31–35 36–40 41–45 46–50 51–55 56–60 61–65 66–70 71–75 76–80 81-85 86–90 >90 Age in years

SM Fruchtman

Survival After Diagnosis in ET

• ET after 10 years – Survival = 72% – Symptom-free survival = 54% • ET after 19 years – Survival = 52% – Symptom-free survival = 35% • In young patients with ET, overall survival in the first 10 years is near normal HS Gilbert

ET: Natural History (2091 Cases)

• 1294 F : 797 M = 1.62

• Mean age 56 years (SD 17, range 2-94) – 20% under age 40 years – 30% 40-60 years of age – 50% over age 60 years • Mean platelets 1.004 million/ m L (range 0.601-4.0) • Splenomegaly 22.8% • Hepatomegaly 27.0% • Karyotypic abnormality 4.9% RM Petitt

ET: Natural History (2091 Cases)

• Median follow-up 4.4 years (mean 5.1, SD 3.9) – 230 thrombotic events (11%); 65 fatal (3.1%) – 130 hemorrhagic events (6.2%); 6 fatal (0.3%) – 137 neoplastic events (6.6%) • 32 acute leukemia (1.5%); 24 fatal (1.2%) • 8 MDS (0.4%); 2 fatal (0.1%) • 31 myelofibrotic (1.5%); 3 fatal (0.15%) • 66 nonhematologic (3.2%); 3 fatal (0.15%) – 201 deaths (9.6%) RM Petitt

Classification of the Erythrocytoses

Raised PCV (Females >0.48; Male >0.51) RCM (interpreted using ICSH reference values)

Increased RCM

Absolute erythrocytosis Primary erythrocytosis

Congenital e.g.

Truncation of the Epo receptor Normal RCM

Apparent erythrocytosis

Acquired e.g.

Polycythemia vera

Secondary erythrocytosis

Congenital e.g.

High oxygen affinity Hb, autonomous high Epo production Acquired e.g.

Hypoxemia, renal disease

Idiopathic erythrocytosis

SM Fruchtman

Clinical Features of P Vera

• Median age at diagnosis = 60 years • Slightly more common in men • Clinical manifestations: thrombosis > hemorrhage; microvascular symptoms (headaches, transient neurologic or ocular symptoms, distal paresthesias, or erythromelalgia) are common • Transformation into myeloid metaplasia occurs in 10% 30% of patients observed for 10 to 25 years; natural evolution or treatment related? • Transformation to MDS and acute leukemia – late complication; natural evolution or treatment related?

HS Gilbert

Thrombosis in P Vera

• Risk of recurrent thrombosis increases with age and in patients with a history of thrombosis: – Reported incidence for thrombotic events ranged from 18%-61%; PVSG incidence was 10% per annum in arm treated with phlebotomy only – Large retrospective study found 41% incidence of thrombosis before diagnosis or during follow-up – 25% risk of recurrent thrombosis in patients with a history of thrombosis; 17% in patients with no risk HS Gilbert

Life Expectancy in P Vera

• In P Vera, median survival exceeds 15 years in young patients.

• In P Vera, serious complications can occur in young patients. Early therapy may be advised.

HS Gilbert

Natural History

Natural History of ET and P Vera

• Hemorrhage • Myeloid metaplasia • Myelofibrosis • Myelodysplastic syndrome • Acute leukemia • Thrombosis HS Gilbert

ET: Complications

• Microvascular occlusion – Erythromelalgia from spontaneous aggregation • Medium/large vessel thrombosis – Probably some baseline risk at any platelet count – May increase somewhat as platelet count rises • Hemorrhage – Risk definitely increases as platelet count rises • Leukemic transformation – Small but definite underlying risk – Effect of treatment RM Petitt

Complications of ET

• Arterial thrombosis (MI 4%, stroke 2%) • Microvascular disturbances (TIAs primarily, erythromelalgia) • Hemorrhage (GI or serious hemorrhage 5%) • Venous thrombosis 20% 32% 18% 6% CM Kessler

Erythromelalgia (Erythermalgia)

Attacks of severe burning, erythema, & warmth (feet > hands) Provoked by heat, exercise, and dependency Relieved by cold, rest, and elevation and by aspirin (?cyclooxygenase role) Two types: Idiopathic (60%): usually symmetrical Secondary (40%): half asymmetric Half of secondary cases associated with chronic MPDs: P Vera in 60%, ET in 40%; MM & CML very rarely Symptom attacks precede overt MPD in 85%, often by several years M:F ratio = 2 Gangrene occurs, especially in patients with ET RM Petitt

Photo

RM Petitt

ET: Risk of Thrombosis

• 114 high-risk ET patients – 97 (85%) over age 60 – 52 (46%) with previous thrombosis – Median platelet count 788,000 (533,000 1,240,000)/μL • Randomized to hydroxyurea (56) or no treatment (58) • After median follow-up of 27 months – 2 thrombotic events (3.6%) in hydroxyurea group – 14 thrombotic events (24%) in untreated group – Statistically significant difference (p = 0.003) RM Petitt

ET: Risk of Thrombosis

• 65 low-risk ET patients – Age<60, no history of thrombosis, platelets<1.5 million/μL (mean 823,000) • Compared with 65 age and sex-matched controls • Median follow-up 4.1 years • Thrombosis incidence: – 1.91 cases per 100 patient years in ET – 1.50 cases per 100 patient years in controls • Authors concluded that low-risk patients do not need treatment. However, this study is subject to criticism because: – Aspirin usage was not controlled – Seven previous studies all had significantly higher rates of thrombosis in their groups of ET patients RM Petitt

ET and Thrombosis

• 56 ET patients 1976-1992 (median age 66, median platelets 1.125 million/ m L at diagnosis, median follow-up 45 months); 52 received platelet-reducing agents; 41 received antiaggregating agents.

• 46 (82%) had symptoms attributable to thrombocythemia – 32 at platelet counts < 600,000/ m L – 23 at platelet counts < 500,000/ m L – 10 at platelet counts < 400,000/ m L • 19 (34%) had severe complications (CVA, TIA, DVT, gangrene) – 10 at platelet counts < 600,000/ m L – 7 at platelet counts < 500,000/ m L – 2 at platelet counts < 400,000/ m L • 42 (91%) improved following further reduction in platelets RM Petitt

Hepatic Venogram of Budd-Chiari Syndrome in ET

CM Kessler

ET: Symptoms and Platelet Count

1000 900 800 700 600 500 400 300 200 100 0 Lowest Counts Associated With Manifestations Counts After Cessation of Manifestations Changes in platelet counts, from the lowest counts recorded while patients were symptomatic to counts recorded on resolution of symptoms in 42 ET patients (4 patients with permanent complications are not included).

RM Petitt

Risk Factors for ET Complications

• Prior thrombotic event • Smoking (31.4% vs 6.6%) (Not well characterized) • Age > 60 years • Long duration of thrombocytosis (platelets > 600,000/ m L —not well characterized) • Other risk factors for CAD (15.1% vs 6.6%) (not well characterized) CM Kessler

Disease Burden of ET and P Vera

Annual incidence • ET • P Vera • AMM 2.5 per 100,000 2.3 per 100,000 1.3 per 100,000 Cases and rate of growth • 93,000 patients – 53,000 ET – 40,000 P Vera • 10% per year growth HS Gilbert

ET P Vera

Timeline of MPDs

MyM MF Leukemia MDS

HS Gilbert

Platelet Physiology

Peripheral Blood

Showing Massive Platelet Clumping and a Basophil

x 1000; Reduced 5% for Reproduction SM Fruchtman

Bone Marrow Biopsy

Showing Prominent Megakaryocytic Hyperplasia

x 100; Reduced 5% for Reproduction SM Fruchtman

Photomicrographs

Showing Variation in Reticulin Content in P Vera Reticulin grade in representative fields of four biopsies: A.

“Normal” B. Slight increase C. Moderate increase D. Marked increase

Foot and Foot Silver Stain.

x250 SM Fruchtman

S S S S SS S S H0 HO Procollagen Molecule OH OH OH Glc Gal OH OH OH OH OH OH OH OH OH OH Gal OH OH OH OH (Man)n GlcNac S S S S S S S S S S S S S S (20 Å) N-Terminal Propeptide (150 Å) S S S S S S S S H0 HO Globular Domain Triple-Helical Domain Nontriple-Helical Domain Collagen Molecule (3000 Å) OH OH OH Glc Gal OH OH OH OH OH OH OH OH OH OH Ga l OH OH OH OH Triple-Helical Domain (15 Å) Nontriple-Helical Domain Nontriple-Helical Domain C-Terminal Propeptide (150 Å) ( Man)n GlcNac S S S S S S S S (100 Å) S S S S S S Schematic representation of the structure of the procollagen molecule.

Glc denotes glucose, Gal galactose, Man mannose, and GlcNac N-acetylglucosamine

SM Fruchtman

What Is the Pathophysiology of Bleeding and Thrombosis in ET

?

• Platelet number • Platelet function • Platelet turnover • Other factors SM Fruchtman

Normal Platelet Physiology

• Platelets participate in hemostasis in conjunction with blood vessels and the humoral coagulation system • Sequence of platelet functions: – Adhesion to exposed subendothelium via vWF and GPIb/IX – Aggregation (cohesion) via fibrinogen interactions with GPIIb/IIIa – Secretion or release reaction with release of dense body and alpha granule contents, FVIII, PAI-1, tPA, etc CM Kessler

Platelet Adhesion

GP IIb/IIIa Endothelium von Willebrand factor Collagen Platelet GP Ia/IIa GP Ib

Adapted from Coller BS. Circulation. 1995; 92: 2373.

CM Kessler

Platelet Aggregation GP IIb/IIIa receptor Platelet Fibrinogen

Adapted from Coller BS. Circulation. 1995;92:2373.

CM Kessler

Platelets at Rest

CM Kessler

Activated Platelets

CM Kessler

Normal Platelet Physiology

• Platelet activation redistributes phosphatidylserine (PS) from the inner leaflet of the platelet membrane to the outer surface; mediated by enzyme phospholipid scramblase • Exposure of PS is critical to expression of platelet procoagulation activity; provide VIIa-TF, Xa, IXa, and VIIIa binding sites CM Kessler

Platelet Structure

CM Kessler

Normal Platelet Physiology (Cont’d )

• Platelet activation induced or modulated by: – Agonist–receptor ligand interaction - activates membrane-bound phospholipases C and A, which hydrolyze phosphatidylinositol on the inner leaflet; mediated by GTP – Inositol triphosphate (IP3) functions as messenger to mobilize Ca +2 ions from intracellular stores – Diacylglycerol (DAG) activates protein kinase C  phosphorylation of 47kD pleckstrin  GPIIb/IIIa activation CM Kessler

Platelet Biochemistry

COX=cyclooxygenase DTS=dense tubular system PKC=protein kinase C DAG=diacylglycerol PIP2=phosphoinositol biphosphate TXA2=thromboxane A2 G-proteins = mediate interaction of cell surface receptors with intracellular efectors CM Kessler

Coagulation Pathway

Intrinsic pathway Extrinsic pathway (Activated within minutes) (Activated within seconds) Contact activation Tissue damage XII XIIa III (Tissue factor) TF VIIa XI XIa, Ca 2 VII IX IXa Ca 2-

ATIII

PF3, Ca+2

TFPI

VIII

PS-PC

VIIIa X Xa V Va Irreversible platelet aggregation

Thrombin (IIa)

Fibrin & blood cells Soluble fibrin XIII XIIIa Ca+2 Cross-linked fibrin clot Fibrinogen Ca 2 PL X Prothrombinase complex Prothrombin (II) KEY Conversion to Causes conversion or release Inhibition of CM Kessler

Functional Abnormalities of Platelets in MPDs

• Prolonged bleeding times - detected in  17% of MPDs; more often seen in AMM than other MPDs; no correlation with bleeding symptoms • Platelet aggregation responses – Spontaneous hyperaggregability in ET – Impaired aggregation to some or all agonists • Impaired epinephrine response most common, but not pathognomonic of MPDs  ADP(39%),  collagen(37%),  epinephrine(57%) CM Kessler

Functional Abnormalities of Platelets in MPDs (Cont’d)

•  platelet  2-adrenergic receptors • Impaired dense granule release • Decreased membrane surface glycoproteins • Defective Ca +2 mobilization,  TxA 2 formation,  lipoxygenase accumulation • Loss of HMW multimers of vWF protein; inversely related to platelet count; improved post cytoreduction CM Kessler

Disorders of Platelet Function

• Clinically manifests as mucocutaneous bleeding, excess hemorrhage posttrauma & surgery, easy bruising, menorrhagia, etc.

• Usually prolonged bleeding time • Platelet aggregation and secretion studies often reveal defect but usually do not predict severity of clinical manifestations • Acquired defects >> inherited defects CM Kessler

Acquired Platelet Dysfunction in MPDs

• Platelets most likely to develop from an abnormal clone of stem cells • Abnormal function may result from enhanced platelet activation in vivo • Platelet abnormalities contribute to the mortality and morbidity of MPD; impact of thrombosis is greater than that of bleeding • Hemorrhagic and thrombotic complications may occur separately or simultaneously; often unpredictable since asymptomatic individuals may have platelet dysfunction CM Kessler

Platelet Disorders in MPDs

• Bleeding and thrombosis less common in CML versus other MPDs • Bleeding more frequent in AMM versus other MPDs; thrombosis not common in AMM • Hemorrhage usually from GI and GU tracts; risks of spontaneous bleeding  with platelets > 2,000,000/ m L; exacerbated by ASA CM Kessler

Platelet Disorders in MPDs (Cont’d)

• Thrombotic events can be arterial and/or venous, often in portal, hepatic, splenic, cerebral sinuses • DVT, PE, AMI, CVA, and PAO all reported in MPDs, often underestimated as platelet mediated events (Underlying etiology is complex; platelet abnormalities may be contributory) • Microvascular arterial thrombi may occur— erythromelalgia, neurologic symptoms, visual complaints CM Kessler

Platelet-Mediated Thrombosis in ET

• Risk factors for thrombosis in ET – Age – Prior thrombotic event – Inadequate control of thrombocytosis – Presence of risk factors for CAD, CVA, etc – In vitro spontaneous megakaryocyte colony formation – Increased risk of recurrent first trimester miscarriages, premature labor, IUFGR (intrauterine fetal growth retardation), abruptio placentae

CM Kessler

Evidence for TPO/Mpl Axis in MPDs

• In 17 patients with ET: – normal to high plasma TPO levels – all patients with strikingly reduced c-Mpl mRNA and protein • normal levels of GPIIb/IIIa – No aberrant pTYR activity in response to TPO stimulation • In 12 patients (10 P Vera, 2 IM): – normal to high plasma TPO levels – all patients with scant or absent c-Mpl protein expression – reduced or absent pTYR activity in response to TPO stimulation • TPO and anagrelide have opposing effects

Observation TPO

Megakaryocyte size Mean ploidy Megakaryocyte colony # Megakaryocyte colony size 50% 32-65 N 7-10 X 5-6 X

Anagrelide

22%  8-16 N  57%  31% J McCarty

Hypothesis:

• Since myeloproliferative disorders are associated with abnormalities of TPO and c-Mpl • Since the biologic effects of TPO and anagrelide appear to act in opposition in vitro • Does anagrelide induce its thrombocytopenic effect by modulating TPO/c-Mpl-specific molecular events in human stem cells?

J McCarty

Consequences of TPO/Mpl Binding

1.

TPO binding dimerizes Mpl on cell surface 2. Causes autophosphorylation of associated JAK/STAT and other pTYR and pSER signaling pathways 3. Activated early acting transcription factors induce transcription of megakaryocyte-specific gene sets 4. Mpl/TPO complex internalized and degraded leading to loss of both receptor and bound TPO 5. Newly synthesized Mpl receptor monomers are refreshed at cell surface Anagrelide may interfere at these steps J McCarty

Effect of Anagrelide on hTPO- or hIL-3-Stimulated Liquid Culture of Human CD34 + Stem Cells 8000 7000 6000 5000 4000 3000 2000 1000 0 12 day stimulation with hTPO 100ng/mL Megakaryocyte number 180000 160000 140000 120000 100000 80000 60000 40000 20000 0 8000 7000 6000 5000 4000 3000 2000 1000 0 12 day stimulation with hIL-3 200U/mL Total mononuclear cell number 180000 160000 140000 120000 100000 80000 60000 40000 20000 0

J McCarty

Summary Findings

• Anagrelide inhibits CFU-Mk in human CD34 + • Antiproliferative effects: – specific for TPO, not IL-3 – specific for megakaryocyte lineage cells – do not affect total MNC number • Anagrelide species-specific effect determined by Mpl • Anagrelide affects pTYR activity in TPO-stimulated, but not IL-3-stimulated, cells and lines • Anagrelide exhibits TPO-specific inhibition of cell proliferation and pTYR activity.

J McCarty

Potential Mechanisms of Anagrelide Action

Anagrelide may act at several steps: 1. May act as inhibitor of TPO binding 2. May inhibit proximal steps of TPO/Mpl-associated phosphotyrosine or phosphoserine signaling pathways 3. May interfere with TPO/Mpl associated megakaryocyte specific gene expression 4. May prevent internalization of bound TPO/Mpl complex 5. May curtail production and cell surface expression of Mpl monomers J McCarty

Diagnosis and Diagnostic Issues

Current Approaches to Diagnosing ET and P Vera

HS Gilbert

ET and P Vera: Initial Considerations

• Diagnosis—confirm • Symptoms at presentation • Assessment of risk—age, risk factors for thrombosis • Discussion of treatment alternatives with patient SM Fruchtman

Diagnosis of P Vera vs ET

• Primary Erythrocytosis – Acquired abnormality of marrow - Polycythemia vera – Congenital abnormality - truncation of erythropoietin receptor • Secondary Erythrocytosis – Acquired - secondary to hypoxemia, renal disease, etc – Abnormalities of the Hgb molecule or autonomous high erythropoietin production • Diagnosis: Measure erythropoietin levels – Elevated hematocrit and low erythropoietin levels – No other symptoms – Patient has P Vera HS Gilbert

Differential Diagnosis of Thrombocytosis

I.

Essential thrombocythemia II. Other chronic myeloproliferative disorders A. Polycythemia vera B. Chronic granulocytic leukemia C. Myelofibrosis/agnogenic myeloid metaplasia D. Overlap myeloproliferative disorders III. Myelodysplastic syndromes associated with thrombocytosis A. 5q-syndrome B. Idiopathic refractory sideroblastic anemia IV. Reactive thrombocytosis A. Blood loss and/or iron deficiency B. Splenectomy C. Hemolytic anemia D. Malignancy E. Myelophthisis F. Chronic inflammatory disorders G. Infection H. Drug induced I. Rebound from thrombocytopenia J. Exercise SM Fruchtman

Proposed Diagnostic Criteria —ET

I.

II.

III.

IV.

V.

Platelet count >600,000/ m L Hematocrit <40, or normal RBC mass (Males <36 mL/kg, females <32 mL/kg) Stainable iron in marrow or normal serum ferritin or normal RBC mean corpuscular volume (otherwise Fe trial) No Philadelphia chromosome or bcr/abl gene rearrangement Collagen fibrosis of marrow A. Absent or B. <1/3 biopsy area without both marked splenomegaly and leukoerythroblastic reaction VI.

No cytogenic or morphologic evidence for MDS VII. No cause for reactive thrombocytosis SM Fruchtman

Thrombosis

• DVT • PE • CVA/TIA/Retinal/MI • Hepatic & portal vein thrombosis • Digital ischemia • Erythromelalgia • Miscarriage SM Fruchtman

Essential Thrombosis

• Most patients are asymptomatic (estimated to be up to 2/3) • However, catastrophic thrombotic complications are seen • Frequently a syndrome of young women. Thus, issues of management during pregnancy are important SM Fruchtman

Mpl and P Vera

Reduced expression of the thrombopoietin receptor Mpl is characteristic of polycythemia vera and idiopathic myelofibrosis.

The abnormality appears to distinguish polycythemia vera from other forms of erythrocytosis.

SM Fruchtman

Bleeding

• Skin • Mucous membranes • Gastrointestinal By trauma SM Fruchtman

Polycythemia Vera Study Group (PVSG) Five Protocols

Polycythemia Vera PVSG-01

Prospective, randomized Phlebotomy vs 32 P vs chlorambucil

PVSG-05

Prospective, randomized Phlebotomy, aspirin, dipyridamole vs 32 P

PVSG-08

Phase II, efficacy Hydroxyurea

Essential Thrombocythemia PVSG-10

Prospective, randomized Melphalan vs 32 P

PVSG-12

Phase II, efficacy Hydroxyurea

# Patients

431 178 51 55 29

Initiated

1967 1977 1977 1975 1977 SM Fruchtman

ET: 1975 PVSG Diagnostic Criteria

• Platelet Count in excess of 1,000,000/ m L • Marked megakaryocytic hyperplasia • Abundant platelet clumps • Normal red cell mass (adequate iron) • No Philadelphia chromosome • No significant myelofibrosis RM Petitt

ET: 1983 PVSG Diagnostic Criteria

• Platelet Count in excess of 600,000/ m L • Marked megakaryocytic hyperplasia • Abundant platelet clumps • Normal red cell mass (adequate iron) • No Philadelphia chromosome • No significant myelofibrosis RM Petitt

ET: 1996 Swedish Diagnostic Criteria

• A1 Platelets in excess of 600,000/ m L • A2 Normal red cell mass, or <125% of mean predicted value with stainable marrow iron or failed iron trial • A3 No Philadelphia chromosome • A4 Megakaryocytic hyperplasia and/or increased ploidy; no fibrosis • B1 Splenomegaly on isotopic scan or ultrasound • B2 Unstimulated growth of BFU-E and/or CFU-Meg • B3 Normal sedimentation rate and fibrinogen • All four “A” criteria = ET • A1 + A2 + A3 + any two “B” criteria = ET RM Petitt

ET: 1997 Revision of 1983 PVSG Criteria

• Platelet count in excess of 600,000/ m L • Hematocrit under 40% or normal red cell mass • Normal marrow iron or serum ferritin or MCV • No Philadelphia chromosome or bcr/abl rearrangement • No collagen fibrosis, or less than 1/3 of biopsy area (without splenomegaly and leukoerythroblastosis) • No cytogenetic or morphologic evidence of MDS • No cause for reactive thrombocytosis RM Petitt

PVSG Criteria for the Diagnosis of P Vera

A1 Increased RBC mass Male:  36 mL/kg Female:  32 mL/kg A2 Normal arterial O 2 (  92%) saturation B1 Thrombocytosis: Platelet count >400,000/ m B2 Leukocytosis: >12,000/ m L (no fever or infection) L A3 Splenomegaly B3 Increased leukocyte alkaline phosphatase (LAP >100) B4 Increased serum B 12 /binders B 12 : (>900 pg/mL) Unbound B 12 binding capacity (>2200 pg/mL) Diagnosis of polycythemia vera virtually certain in the presence of A1+A2+A3 or A1+A2+ any two from category B.

SM Fruchtman

Diagnostic and Confirmative Criteria of P Vera Proposed by the PVSG

A Diagnostic criteria

A1 Raised red cell mass male >36 mL/kg female >32 mL/kg A2 Absence of any cause of secondary erythrocytosis by clinical and laboratory investigations A3 Histopathology of bone marrow by biopsy a. increase of celluarity, panmyelosis b. increase and clusters of enlarged megakaryocytes with hyperploid nuclei c. reticulin fibers (optional)

B Confirmative criteria

B1 Thrombocythemia platelet count >400,000/ isotope/ultrasound scan m L B2 Granulocytes >100,000/ m L and/or raised neutrophil alkaline phosphatase score in the absence of fever or injection B3 Splenomegaly on palpation or B4 Erythroid colony formation in absence of EPO: spontaneous EEC A1+A2+A3 is consistent with early stage P Vera (so called “idiopathic erythrocytosis”) A1+A2+A3 plus any from the category B establishes overt P Vera A3+B1 is consistent with essential thrombocythemia A3+B3 and/or B4 is consistent with a primary myeloproliferative disorder SM Fruchtman

Proposed Diagnostic Criteria for P Vera

A1 Raised red cell mass (>25% above mean normal predicted value) A2 Absence of cause of secondary polycythemia A3 Palpable splenomegaly A4 Clonality marker e.g. Abnormal marrow karyotype A1+A2+A3 or A4 establishes P Vera A1+A2+two of B establishes P Vera B1 Thrombocytosis (platelet count >400,000/ m L) B2 Neutrophil leukocytosis (neutrophil count >10,000/ m L) B3 Splenomegaly demonstrated on isotope/ultrasound scanning B4 Characteristic BFU-E growth or reduced serum erythropoietin SM Fruchtman

ET: 1999 British Diagnostic Criteria

• Platelets exceeding 400,000/ m L (600,000/ m L for trials) • No cause for secondary thrombocytosis (congenital or acquired) • No other cause for myeloproliferative thrombocythemia (P Vera, CML, AMM-MF, MDS) • Compatible marrow histology – Increased, clustered large megakaryocytes – Hyperlobulated megakaryocyte nuclei – No collagen fibrosis or osteosclerosis RM Petitt

ET: 1999 Dutch Diagnostic Criteria

• Diagnostic Criteria: – A1 Platelets exceeding 400,000/ m L without reactive cause – A2 Increased, clustered, mature giant megakaryocytes with hyperploid nuclei – A3 No preceding myeloproliferative or myelodysplastic disease • Confirmation Criteria: – B1 – B2 Normal or elevated LAP score, normal ESR, no fever Marrow cellularity normal or slightly increased; no or minimal reticulin fibrosis – B3 – B4 Palpable splenomegaly, or >11 cm on U/S or CT Spontaneous erythroid and/or megakaryocyte colony growth RM Petitt

2002 WHO Classification of Chronic MPDs

Chronic myelogenous leukemia (bcr/abl +) Chronic neutrophilic leukemia Chronic eosinophilic leukemia Polycythemia vera Chronic idiopathic myelofibrosis Essential thrombocythemia Chronic myeloproliferative disease, unclassifiable RM Petitt

Survival Time (%) for Patients in Various Treatment Groups

100 Various combinations with X-ray therapy X-ray therapy 32 P and/or chlorambucil Venesection Untreated 50 5 2 4 6 8 10 12 Years of duration of the disease 14 16

SM Fruchtman

Cumulative Survival on Study

1.0

0.8

0.6

0.4

0.2

0.0

0 Legend Phlebotomy Chlorambucil 32 P Total 134 141 156 Events Breslow X 2 2 = 4.394 p=.1111

54 82 83 Logrank X 2 2 = 9.650 p=.0080

100 200 300 400 500 Time in weeks 600 700 800

SM Fruchtman

Cumulative Survival After 7 Years on Study

1.0

0.8

0.6

0.4

0.2

0.0

0 Legend Phlebotomy Total 59 Chlorambucil 63 32 P 92 Events 16 35 41 Breslow X 2 2 = 16.968 p=.0002 Logrank X 2 2 = 16.465

p=.0003

100 200 300 400 Time in weeks after 7 years 500 600

SM Fruchtman

PVSG-01: Thrombosis (On Study Events) 1/1/86

1.0

0.9

0.8

0.7

0.6

0.5

0 100 Comparison Statistics Breslow Logrank X 2 2 8.54

4.89

p

.01

.09

200 Legend Phlebotomy Chlorambucil 32 P Total 134 141 156 Events 51 41 51 300 400 500 Time in weeks 600 700 800

SM Fruchtman

Causes of Death by Treatment Group

Number at start Number of deaths

Causes of death

Thrombosis Hemorrhage Leukemia/Lymphoma Cancer Spent/MF Other

Phlebotomy

134 53

CLB

141 82

Percent

17.2

0.0

1.5

4.5

0.7

15.7

13.5

4.3

20.6

9.9

2.1

7.8

32 P

156 79 13.5

3.8

11.5

9.6

1.3

10.9

Total

431 214 44.2

8.1

33.6

24.0

4.1

34.4

SM Fruchtman

1.00

.80

Survival Probability in Patients > 70 years

Age >70 32 P Chlorambucil .60

Phlebotomy .40

.20

0 0 50 100 150 200 250 300 Weeks since randomization 350 400

SM Fruchtman

PVSG-01: Initial Thrombotic Event (On Study Events)

Type of event

Cerebral vascular accident Myocardial infarction Peripheral arterial occlusion Pulmonary infarction Venous thrombosis (other than thrombophlebitis) Deep vein thrombophlebitis Miscellaneous Total

Number

51 18 13 8 6 32 18 146

% of events

35 12 9 6 4 22 12 100 SM Fruchtman

PVSG-01: Thrombosis-Related Risk Factors

• Multivariate Analysis – Randomization to phlebotomy – Age >70 years – Previous thrombosis SM Fruchtman

PVSG-01: Matched Pair Analysis

For each patient with thrombosis, a thrombosis-free control patient of similar age, sex, treatment group, and time on study was selected.

Most recent hematocrits and platelet counts were not different. Hematocrit over 52% and platelet counts over 1,500,000/ m L were rare.

SM Fruchtman

Hematologic Parameters and Relative Odds of Thrombosis:

Matched Pairs Case-Control Study Estimated odds of thrombosis p-value* Hematologic parameter Platelets (x10 3 /

m

L)

 1,000 vs <1,000  600 vs <600  450 vs <450

Hematocrit (%)

 52 vs <52  47 vs <47 >45 vs  45 *From Mantel-Haenszel Chi-Square.

0.7

1.4

0.9

1.3

1.2

1.1

>0.9

>0.5

>0.9

>0.5

>0.5

>0.9

SM Fruchtman

Risk Factors Associated with Thrombosis Cox-Model Multivariate Analysis

Risk factor

Phlebotomy vs 32 P Chlorambucil vs 32 P History of thrombosis

431 Patients Estimated relative risk

1.7

0.8

2.1

T*

2.3

-0.7

3.3

Test of model

X 2 4 =43.0

p

<.001

Age (70 vs 50) 2.3

4.5

T*  1.96 indicated significant association with incidence of thrombosis (

p

 0.05) SM Fruchtman

Age-Specific Therapy for P Vera

Age

>70 years 50 –70 years <50 years

Initial choice of therapy

32 P Phlebotomy (Hydroxyurea, 32 P) Phlebotomy (Hydroxyurea) SM Fruchtman

Survival

On study

Median time to death (yrs) Maximum follow-up (yrs)

Phlebotomy CLB

12.5

8.9

17.5

18.2

32 P

11.4

16.6

SM Fruchtman

PVSG-08

Polycythemia Vera Prior myelosuppressive Rx (RT or Chemo) No prior Rx or Hydroxyurea 30 mg/kg/day One Week Hydroxyurea 15 mg/kg/day and adjusted as needed phlebotomy

SM Fruchtman

Clinical and Laboratory Characteristics at Initial Evaluation by Treatment and Protocol

Evaluation Prior thrombosis % Hematocrit % RBC X 10 8 / m L Platelets X 10 3 / m L <600,000/ m L 600,000-1,000,000/ m L >1,000,000/ m L

PVSG-01 Phlebotomy

134 14.2

61.7

±7.5

7.16

±1.09

505 ±24.6

76 17 7

PVSG-08 Hydroxyurea

51 35.3

52.9

±8.1

6.60

±0.86

778 ±63.6

47 31 22 SM Fruchtman

Sex and Age at Initial Evaluation by Treatment and Protocol

Number Male (%) <50 years 50 –70 years  70 years Female (%) <50 years 50 –70 years  70 years

PVSG-01 Phlebotomy

134 54.5

11.2

31.3

11.9

45.5

9.7

25.4

10.4

PVSG-08 Hydroxyurea

51 52.9

11.8

31.4

9.8

47.1

15.7

25.5

5.9

SM Fruchtman

Comparative Incidence of Thrombosis by Year by Protocol and Treatment Group Cumulative % Year 1 Year 2 Protocol

A. On study events HU (PVSG-08 NPT) Phlebotomy (PVSG-01) B. All events HU (PVSG-08 NPT) Phlebotomy (PVSG-01) 2.8

±4.0

8.7

±2.5

5.9

±3.3

9.0

±2.5

6.6

±3.7

14.0

±3.2

7.9

±3.8

15.8

±3.2

SM Fruchtman

Comparative Incidence of Thrombosis by Protocol and Treatment Group

(On Study, First 378 Weeks) Protocol

HU (PVSG-08 NPT)

Total patients

51

# Events

5

%

9.8

44 32.8

Phlebotomy (PVSG-01) Wilcoxon X 2 1 =5.58

Logrank X 2 1 =6.80

134

p

=.018

p

=.009

SM Fruchtman

Comparative Incidence of Acute Leukemia by Protocol and Treatment

On study events, first 795 weeks of study

Protocol

HU-NPT (PVSG-08) Phlebotomy (PVSG-01) Wilcoxon X 2 1 =1.305

Logrank X 2 1 =1.791

All events HU-NPT (PVSG-08) Phlebotomy (PVSG-01) Wilcoxon X 2 1 =2.749

Logrank X 2 1 =2.344

Patients

51 134

p

=.2532

p

=.1808

51 134

p

=.0973

p

=.1258

Total # events

3 2 5 5

%

5.9

1.5

9.8

3.7

SM Fruchtman

PVSG-01: Leukemia by Treatment

Protocol Total patients

On study events, first 795 weeks of study

# events

CHL 141 24 32 P Phlebotomy Wilcoxon X 2 1 =14.718

Logrank X 2 1 =20.938

156 134

p

 .0001

p

 .0001

17 2

%

17.0

10.9

1.5

SM Fruchtman

Survival by Prior Therapy and Protocol

On study events, first 795 weeks of study

Protocol

HU-NPT (PVSG-08) Phlebotomy (PVSG-01) Wilcoxon X 2 1 =3.241

Logrank X 2 1 =2.227

All events HU-NPT (PVSG-08) Phlebotomy (PVSG-01) Wilcoxon X 2 1 =2.300

Logrank X 2 1 =1.847

Patients

51 134

p

=.0718

p

=.1356

p p

51 134 =.1293

=.1742

Total # events

16 54

%

31.4

40.3

20 74 39.2

55.2

SM Fruchtman

Risk of Leukemic Transformation with Myelosuppressive Therapy

Therapy

No treatment or aspirin Phlebotomy Hydroxyurea 32 P Alkylating agent More than 1 myelosuppressive agent IFN 

Polycythemia vera (%)

– 2/18 (11) 1/16 (6) 0/2 – 1/4 (25) 0/2

P Vera, MF, ET

0/11 2/18 2/25 0/2 0/1 1/5 0/2 SM Fruchtman

Eligibility

PVSG-05

1. No previous treatment except phlebotomy 2. Disease diagnosed within four years 3. Fulfillment of diagnostic criteria 4. No chronic disorder requiring long-term ASA Phlebotomy to HCT  40% Randomization

Phlebotomy Prn to maintain Hct <45% ASA 300 mg TID Dipyridamole 75 mg TID 32 P 2.7 mc/M 2 i.v.

Q 12 wks prn (limit 5 mc/dose) Phlebotomy for Hct >45% Increase dose by 25% if no response

SM Fruchtman

PVSG-05: Thrombosis and Hemorrhage-Free Cumulative Survival

1.00

0.95

0.90

0.85

0 Legend Treatment Total in Group Events Phleb/Persantine/ASA 88 7 P-32 90 2 10 Comparison Statistics Breslow X 2 2 2.8

p

.09

Logrank 3.1

.08

20 30 40 50 60 70 Time in weeks 80 90 100 110

SM Fruchtman

PVSG-05: Thrombosis and Hemorrhage-Free Cumulative Survival

1.00

95 90 85 80 0 Legend Treatment Total in Group Events Phleb/Persantine/ASA 88 13 P-32 90 2 10 20 30 40 50 60 70 Time in weeks 80 90 100 110 Comparison Statistics Breslow X 2 2 8.660

P

0.0033

Logrank 9.018

0.0027

SM Fruchtman

Summary of 91 Patients with Thrombocytosis Treated with PVSG Protocols Treatment protocol

HU only 32 P only Melphalan only Melphalan + HU 32 P + HU HU + other Melphalan + 32 P Melphalan + 32 P + HU No treatment Total study population

Median follow-up (years)

8.2

5.6

4.4

10.5

8.4

7.7

4.5

9.2

4.4

7.0

Total patients

22 16 14 10 9 7 4 2 7 91

No. AML (all dead)

1 2 1* 0 1 5 1* 0 1 12

No. died of other causes

7 5 6 4 2 1 2 0 2 29

No. alive at last follow-up

14 9 7 6 6 1 1 2 4 50 *AMLs in patients in ET [?MF] category.

SM Fruchtman

The Mount Sinai Myeloproliferative Study

Jan 1986 –Dec 1998 Disorder studied

P Vera ET MM/MF

# of patients

167

# of specimens

234 40 63 40 90

# of technically inadequate specimens

23.4

(10%) 8.0

(20%) 18.90

(20%)

# of evaluable patients

156 32 56 SM Fruchtman

The Mount Sinai Myeloproliferative Study

Jan 1986 –Dec 1998 Disorder

P Vera

# of patients

156

Normal karyotype

109 69.8%

Normal with nonclonal abnormalities

5 3.2%

Normal at Dx/ Abnormal on follow-up

5 3.2%

Abnormal at Dx

37 23.7% ET 32 0 MM/MF 56 27 84.3% 30 53.5% 1 3.1% 0 0 0 0 4 12.5% 26 46.4% SM Fruchtman

Frequency of Chromosomal Abnormalities

Abnormality

del (20) (q11q12) +8 +9 del (7q) -7/t (7p) del (9q)/i (9q) abnormalities 1p t (16q) del (13)(q12-13q14-21) -5

% of total P Vera % of abnormal

4.4

3.8

3.2

3.2

18.9

16.2

13.5

13.5

0 2.5

1.9

1.9

0 0 0 10.8

8.1

8.1

0 0

% of total MM/MF % of abnormal

5.3

1.7

0 0 11.5

3.8

0 0 10.7

0 7.1

0 12.5

5.3

23 0 0 0 26.9

11.5

SM Fruchtman

Treatment

Treatment: Essential Thrombocythemia

Treatment Goals for ET

• No therapy to eliminate malignant stem cell clone • Direct therapy at reducing mortality and life-threatening complications • In high-risk ET, reduce platelets  600,000/µL; ?  450,000/ m L • No adequate prospective, randomized, controlled studies CM Kessler

Conditions Suggesting Urgent Treatment

• Symptomatic coronary artery disease • Transient ischemic attacks • Major hemorrhage • Active thrombosis/pulmonary embolism HS Gilbert

Urgent Treatment of ET

• Platelet apheresis - typically 1-3 treatments with target platelet count < 400,000/ m L • Hydroxyurea - target platelet count < 400,000/ m L • Nitrogen mustard, 0.4 mg/kg if apheresis not available HS Gilbert

ET: Prognostic Factors (2091 Cases)

• Favorable factors – Platelets less than 700,000/ m L – Treatment with antiaggregating or cytostatic drugs • Unfavorable factors – Previous thrombosis – Older patients – Platelets exceeding 1.5 million/ m L at diagnosis – Platelets exceeding 1.0 million/ m L at follow-up RM Petitt

Treatment of ET

There is evidence that ET is a heterogeneous disorder both clinically and on a stem/progenitor cell level.

Thus, the dilemma exists as to when to employ agents that either lower the platelet number and/or inhibit platelet function.

SM Fruchtman

Risk-Based Treatment for ET

• Low risk – Age < 60 years – Platelets < 1,500,000/ m L – No history of thrombosis – No cardiovascular risks • No treatment or low-dose aspirin HS Gilbert

Risk-Based Treatment for ET

• Intermediate risk – Age < 60 years – No history of thrombosis – Platelet counts > 1,500,000/ m L or – Cardiovascular risk factors HS Gilbert

Risk-Based Treatment for ET

• Intermediate-risk treatment – Treat cardiovascular risk factors – Avoid aspirin if platelet counts > 1,500,000/ m L – No treatment or anagrelide; hydroxyurea or IFN  HS Gilbert

ET: The High-Risk Patient

• Age 60 or greater • Previous thrombosis (or comorbid disposition such as DM, PVD, HTN or thrombophilia) • Platelets exceeding 1.0 or 1.5 million/ m L • Peripheral myeloid immaturity RM Petitt

Risk-Based Treatment for ET

• High risk – Age  60 years or History of thrombosis • • Hydroxyurea and low-dose aspirin; anagrelide IFN  , 32 P, busulfan, pipobroman HS Gilbert

ET: The Risks of Observation

• Thrombosis – A baseline risk which does not increase significantly as platelets rise to exceedingly high levels – Risk aggravated by unrecognized thrombophilic states • Hemorrhage – Risk increases significantly as platelets rise above 1.5 million/ m L – Acquired vonWillebrand disease with high platelet counts • Leukemic transformation – All chronic myeloproliferative disorders have an underlying risk of transformation to acute leukemia. This risk is independent of any treatment and is estimated to be 90% in CML, 7%-10% in MM-MF, 1.5% in P Vera, and 1%-2% in ET RM Petitt

Risk of Elevated Platelets in ET

• Is there clear evidence that elevated platelet levels are bad?

– In symptomatic patients?

– In asymptomatic patients with very high platelet counts?

– In patients with mild to moderate thrombocythemia?

– What is the role of plateletpheresis?

CM Kessler

Risk of Elevated Platelets: New Data

• Regev et al: Thrombotic complications not uncommon in ET, even at low platelet levels. Recommendation: Symptomatic patients with relatively low platelet counts be treated and platelets be reduced to low-normal range • Storen, Tefferi: Long-term anagrelide study. All thrombohemorrhagic complications occurred at platelet counts of > 400,000/ m L. Normalization of platelets may be required to minimize risk CM Kessler

ET: The Risks of Treatment

Treatment-related adverse events reflect agent(s) chosen, dose and duration of therapy: • Leukemic transformation • Bone marrow suppression • Less severe cardiac, dermatologic, gastrointestinal and neurologic side effects RM Petitt

ET: Treatment

• Antiaggregating agents • Platelet apheresis • Myelosuppressive agents – Alkylating agents – Radiophosphorus – Hydroxyurea • Maturation Modulators – IFN  2A, recombinant – Anagrelide • Bone marrow transplantation • Combinations of above RM Petitt

Aspirin

• Suppression of thromboxane biosynthesis, which is increased in P Vera and ET patients, can be achieved with low-dose aspirin (~50 mg/day) • Prevention of thrombosis? Under study • Control of microvascular symptoms at low dose in patients without a bleeding diathesis SM Fruchtman

Busulfan

• Busulfan in ET – WEEK 1 - 4 mg per day – WEEKS 2, 3 and 4 - 2 mg per day – TO RESPONSE - 2 mg every other day • Median cumulative dose - 124 mg and duration 3 months HS Gilbert

ET and Busulfan Treatment

• 75 patients (mean age 48; F:M = 1.5) – Splenomegaly 49% – Hepatomegaly 38% – Thrombosis 29% – Hemorrhage 19% • 64 (83%) treated with busulfan – Mean follow-up 9.2 years – 53% died – 18% developed leukemia – 19% developed thrombosis RM Petitt

Busulfan

• Myleran, registered trademark of Glaxo Wellcome • 2 mg tablets • Bifunctional alkylating agent (not a nitrogen mustard analog) • 2003 Physicians’ Desk Reference,* p. 1595: “MYLERAN is contraindicated in patients in whom a definitive diagnosis of chronic myelogenous leukemia has not been firmly established.” *copyright 2003 by Medical Economics Company, Inc., Montvale, NJ RM Petitt

Hydroxyurea (HU)

•  Risk of thrombosis: Hydroxyurea reduced the risk of thrombosis from 24% to <4% in a randomized study of high-risk ET patients • Leukemic conversion: Several nonrandomized studies have supported or refuted a significant increase in leukemic conversion with long-term use of hydroxyurea – In ET, rates range from 0%-5.5% – In P Vera, rates range from 2.1%-10% SM Fruchtman

ET and Hydroxyurea Treatment

• 75 high-risk patients (mean age 62) – Treated with hydroxyurea – Mean follow-up 6.9 years – 6 (8%) developed leukemia • Mean age 65 (35-75) • Mean hydroxyurea exposure 7.2 years • 20 low-risk patients – No treatment (some on aspirin) – No leukemia RM Petitt

ET and Hydroxyurea Treatment

• 114 randomized high-risk ET (56 HU, 58 no Rx) – 15 on busulfan before randomization – 13 on hydroxyurea before randomization – 29 controls changed to hydroxyurea during study (26 thrombosis) • Original follow-up median 27 months. After 73 months: – Survival same; thrombosis rate 9% vs 45% (

p

= <0.0001) • Conclusion: – Sequential use of busulfan and hydroxyurea carries increased risk of second malignancies • One cancer (breast) in untreated group • Seven in hydroxyurea group (5 hematologic:2 ANLL, 2 MDS, 1 CLL) RM Petitt

Hydroxyurea for Patients with ET and a High Risk of Thrombosis

• Prospective randomized trial of 114 patients with ET – Age >60 years or – Previous thrombosis or both – Platelet count <1,500,000/ m L (uncomfortable with randomization) • Randomization – HU vs control (no HU) – 70% were already on antiplatelet agents SM Fruchtman

Probability of Thrombosis-Free Survival

in 114 Patients with Essential Thrombocythemia Treated with Hydroxyurea or Left Untreated 1200 1000 800 Control (n=58) Hydroxyurea (n=56) 600 400 0 6 12 18 Months 24 30 36

SM Fruchtman

Probability of Thrombosis-Free Survival

in 114 Patients with Essential Thrombocythemia Treated with Hydroxyurea or Left Untreated 100 80 60 40 Hydroxyurea (n=56) Control (n=58) 20 p=0.005

0 0 6 12 18 24 30 36 Months after randomization 42 48

The

p

value is for the difference between the two groups (by the log-rank test). The median follow-up was 27 months. Tick marks indicate surviving patients who were continuously free of thrombosis.

SM Fruchtman

Thrombosis in 114 Patients with ET

Type of thrombosis

Arterial Transient ischemic attacks Digital microvascular ischemia Stroke Myocardial infarction Venous Superficial thrombophlebitis Ileofemoral venous thrombosis Total (% of treatment group)

Hydroxyurea group (N=56)

no. (%)

Control group (N=58)

2 (100) 0 0 11 (79) 5 5 1 1 0 0 0 2 (3.6) 1 0 3 (21) 2 1 14 (24)* *There was a difference of 20.4 percentage points in the rate of thrombosis between the groups (95 percent confidence interval, 8.5 to 32 percent; chi square with Yates’ correction, 8.3; 1 df;

p

=0.003).

SM Fruchtman

Hydroxyurea: Adverse Reactions

• Hematologic – Bone marrow depression (leukopenia, anemia, thrombocytopenia) • Gastrointestinal – Stomatitis, anorexia, nausea, vomiting, diarrhea, constipation • Dermatologic – Rash, painful ulceration, erythema, hyperpigmentation, squamous cell skin cancers, dermatomyositis-like changes • Neurologic – Headache, dizziness, hallucinations, convulsions (all rare) • Systemic – Fever, chills, malaise, asthenia RM Petitt

Photo

HS Gilbert

Hydroxyurea and Acute Leukemia in MPDs

50 consecutive MPD patients (30 P Vera, 10 ET, 10 MF) were treated with hydroxyurea (HU), largely for high platelet counts or symptomatic splenomegaly.

9 (18% of total) developed ANLL, and 1 MDS 7/9 (14% of total; 3 P Vera, 1 ET, 3 MF) were treated solely with hydroxyurea average time from CMPD to ANLL = 6.3 years average duration of HU treatment = 3.9 years RM Petitt

Evolution to AML and MDS By Cytoreductive Agent in 357 ET Patients No. of ET patients treated Cases of AML or MDS No. (%) Agent

32 P Alone And other agents* Total Busulfan Alone And other agents* Total HU Alone And other agents † Total Pipobroman Alone And other agents* Total Untreated 29 11 40 35 6 41 201 50 251 12 31 43 31 2 (7) 1 (9) 3 (7.5) 1 (3) 1 (17) 2 (5) 7 (3.5) 7 (14) 14 (5.5) 0 (0) 5 (16) 5 (12) 0 (0) *Generally HU; † Generally pipobroman.

SM Fruchtman

ET and AML/MDS

Median follow-up 98 months (12 yrs) Total 17; AML (6) or MDS (11) 7 HU & other agents (pipobroman, 5; 32 P, 1; Busulfan, 1) 7 HU alone (3.5%) 2 32 P 1 Busulfan Median interval between dx of ET & dx AML/MDS 84 months 7 Patients with AML/MDS & 17P deletion received HU 3/7 received only HU SM Fruchtman

Acquired DNA Mutations and HU

Patient Population

Adults with SCD No HU exposure Short HU exposure Children with SCD Short HU exposure Longer HU exposure Adults with MPD Low HU exposure Prolonged HU exposure Normal controls

# of patients

30 15 15 34 17 17 27 15 12 32

Age in years (mean ± 1SD)

27 ±12 29 ±9 11 ±3 13 ±3 57 ±17 62 ±16 43 ±15

Median HU exposure

None 24 months 7 months 30 months 0 months 11 years None SM Fruchtman

Acquired DNA Mutations and HU (Cont’d)

Patient population

Adults with SCD No HU exposure Short HU exposure Children with SCD Short HU exposure Longer HU exposure Adults with MPD Low HU exposure Prolonged HU exposure Normal adult controls

HPRT assay CE (%) M L X 10 -6

15.1

±12.3

12.4

±8.2

19.1

±19.1

16.7

±10.9

VDJ assay Events per

m

g DNA

1.07

1.14

±0.38

±0.38

13.2

±6.1

20.9

±10.2

11.2

±6.7

9.2

±7.8

1.58

±0.87

1.82

±1.20

12.8

±8.9

12.2

±8.4

16.0

±8.7

37.3

41.1

25.8

±37.6

±29.3

±24.8

1.06

0.64

1.04

±0.73

±0.29

±0.38

SM Fruchtman

Treatment

Thrombotic Complications

In 68 Patients with ET Who Had Long-Term Follow-up, According to Treatment Strategy

Watchful waiting

Thrombotic Events

No.

27

Duration of Follow-up

Person-yr 127

Incidence

Events/100 person-yr 21.3

Low-dose aspirin Cytoreduction † 5 10 Low-dose aspirin and cytoreduction 0 *

p

<0.001 (X 2 =17.3, 1 df) for the comparison with watchful waiting.

† Denotes treatment with busulfan or hydroxyurea.

p

<0.02 (X 2 =6.0) for the comparison with watchful waiting.

§

p

<0.02 (X 2 =8.6) for the comparison with watchful waiting.

139 113 40 3.6* 8.9

‡ 0 § CM Kessler

ET and Interferon Treatment

• Antiproliferative effect on CFU-MK and CFU-GEMM – Nonleukemogenic, nongonadotoxic • Usual daily doses 1–5 million units daily, 3-7 x weekly – Gradual response: peripheral 1-3 months, marrow 9-12 months • 26 ET patients – Median age 48 – Dose-limiting side effects in 24% – 88% responded (62% CR, 26% PR) – Responders crossed over to pipobroman after 12 months RM Petitt

IFN-

In essential thrombocythemia

• Produces platelet reduction (80%-100% response rate), resolution of splenomegaly, and control of disease-associated symptoms • May be used in high-risk women with ET of childbearing age or those who are pregnant • ~20% of patients may not tolerate IFN because of side effects (esp. flu-like symptoms, fatigue, nausea, depression, fever, chills, arthralgias, autoimmune disorders). Higher rates in elderly individuals SM Fruchtman

Photo

HS Gilbert

Management of ET During Pregnancy

Options 1. Observe —? Medical–legal issues 2. ASA 3. ASA & heparin 4. IFN  5. Platelet apheresis for emergencies SM Fruchtman

Women of Childbearing Age with ET

• High risk – IFN  and low-dose aspirin • All others – No treatment or low-dose aspirin HS Gilbert

Anagrelide

For thrombocytosis accompanying any MPD • Indicated for the treatment of patients with thrombocythemia secondary to MPDs to reduce the elevated platelet count and risk of thrombosis and to ameliorate associated symptoms, including thrombohemorrhagic events SM Fruchtman

Anagrelide: Observed Effects

• Reduces platelet count • Reduces platelet turnover rate (elevated in ET) • May reduce megakaryocyte number • Reduces megakaryocyte diameter • Reduces megakaryocyte volume • Normalizes megakaryocyte ploidy

Anagrelide reduces megakaryocytic hyperproliferation and megakaryocytic differentiation

RM Petitt

Anagrelide

• Starting dose - 0.5 mg, orally, 4 times per day • OK to cross over with hydroxyurea • 15% of patients will discontinue • Manufacturer’s target < 600,000/ m L; our target < 400,000/ m L HS Gilbert

ET Patient Response to Anagrelide

• Platelet counts lowered in 7 to 14 days • Sustained response in patients followed for up to 4 years (

p

<.001 vs baseline) • Response seen in 242 patients who had failed previous therapy to reduce platelets

82% Overall Response Rate* (p<0.05) 88% Overall Reduction in ET-Related Symptoms (p<0.05)

CM Kessler

ET: Reduction in Serious Complications with Anagrelide

35

• Reported serious complications of thrombocythemia decreased by 82% after 235 patients took anagrelide for 4 months

30 25 20 15 Preinfarction angina GI bleeds TIAs 10 5 1-4 5-8 9-12 Months on anagrelide therapy

CM Kessler

Anagrelide: Precise & Specific

• Dose-related reduction of platelet production resulting from a decrease in megakaryocyte hypermaturation • Clinically insignificant effect on white blood cells • A slight reduction (~5%) in Hb and PCV occurs with prolonged anagrelide therapy SM Fruchtman

Anagrelide: Precise & Specific (Cont’d)

• No effect on DNA synthesis – No apparent association with oncogenesis – No apparent leukemogenicity SM Fruchtman

Anagrelide

How effective is it?

• Only 19% of patients failed to respond (had <20% reduction in platelet count) in a population that included a large number of nonresponders to other agents. HS Gilbert

Anagrelide

What is unique about it?

• It is the only cytoreductive agent that has specificity for platelets. Therapy targeted at platelets spares the red blood cells and neutrophils in patients who have normal or reduced circulating levels of these cells.

HS Gilbert

Anagrelide

What is its safety and toxicity profile?

• In the analysis of 577 patients, the drop out rate from toxicity was 16% in over 5 years of experience. In the analysis of 942 patients, there were no deaths attributable to anagrelide.

HS Gilbert

Anagrelide: Adverse Reactions

161 of the original 942 patients (17.1%) stopped anagrelide treatment, citing the following reasons: Headache Diarrhea Edema Palpitations Abdominal Discomfort 40 (25%) 19 (12%) 18 (11%) 17 (11%) 12 (7%) (4.2% of all treated) (2.0% of all treated) (1.9% of all treated) (1.8% of all treated) (1.3% of all treated) RM Petitt

Adverse Reactions Leading to Discontinuation of Anagrelide

Type of Adverse Reaction in 577 patients Neurologic (headache, confusion) Gastrointestinal (nausea, abdominal pain, diarrhea) Cardiac (congestive heart failure, edema) Pulmonary Other Number (%) Discontinued 30 (21) 25 (28) 23 (21) 2 (1) 14 (11) HS Gilbert

Anagrelide Study Patients

90% of the original study group of 942 patients had already received other treatment for their myeloproliferative disorders: Hydroxyurea Alkylating Agents IFN Radiophosphorus Other 651 198 94 66 27 (67%) (21%) (10%) (7%) (3%) RM Petitt

Long-term Experience with Anagrelide in Young ET Patients

• Retrospective series of 35 patients (ages 17-48 years) who received anagrelide before 1992 • 82% of 33 responding patients remained on anagrelide for a median of 10.8 years • Of these, 66% were CRs, 34% were PRs CM Kessler

Long-term Experience with Anagrelide in Young ET Patients (Cont’d)

• All thrombohemorrhagic complications occurred at a platelet count of > 400,000/ m L • Complete normalization of platelet counts may be required to minimize residual hemorrhagic risk during therapy • Initial side effects decreased with time; long-term treatment was associated with mild to moderate anemia CM Kessler

Contraindications to Anagrelide

• Uncontrolled congestive heart failure – Aggravation by additional fluid retention • Pregnancy – Fetus at risk for transplacental drug exposure • Lactation – Infant at risk for drug ingestion RM Petitt

Anagrelide Maintenance Doses (mg/day)

6 mos.

12 mos.

18 mos.

P Vera ET CML OMPD 2.8

2.4

2.3

2.1

2.8

2.5

2.1

1.8

2.8

2.3

2.1

1.7

RM Petitt

Long-term Anagrelide Treatment

40 patients started on treatment 1985-1988: 4 nonresponders 2 lost to follow-up 9 deaths 3 blast transformation of CML 2 colon cancer 2 myocardial infarction 2 unknown 25 evaluable after 10 or more years of treatment RM Petitt

Long-term Anagrelide Treatment

25 older patients with chronic myeloproliferative disorders taking anagrelide for 15 or more years: 6 developed mild renal insufficiency (creatinines 2-3) all poorly controlled hypertensives azotemia appeared 3-11 years after starting maintenance dose decreased as creatinine rose 19 have had no change in dosage or response 34 young women treated with anagrelide for >10 years: no changes in renal function RM Petitt

Anagrelide Study 301: Overview Safety 3,660 Efficacy 1,618 2251

ET

462

P Vera (12.6%)

1409

Non-ET

954

CML, MF, OMPD

934

ET

208

P Vera (12.9%)

684

Non-ET

476

CML,MF, OMPD

SM Fruchtman

Anagrelide Study 301: Response Definitions

Complete response Platelets <600,000/ m L or  50% reduction from baseline at least 4 weeks after starting anagrelide therapy Partial response 20%-50% reduction from baseline SM Fruchtman

Anagrelide Study 301: Response (Efficacy Population)

120 100 80 60 40 20 0 100 63.7

11.5

75.7

All Patients Complete Response Partial Response Total Response

SM Fruchtman

Anagrelide Study 301: Response in Patients with Platelets

1,500,000/

m

L

n Total Response (%) Median time to response (days) (95% CI) ET Patients

116

Non-ET

158 99 (85.3) 125 (79.1) 64 (53-76) 63 (54-81) SM Fruchtman

Anagrelide Study 301: Transformation of ET Patients (n=2251)

n AL transformation Non-transformed 47 2180 †

22 additional patients had other transformations and 2 patients were misdiagnosed Median cumulative dose (mg) 342* 746*

*

p=0.005

SM Fruchtman

Anagrelide Study 301: Safety Conclusions

1. Essential thrombocythemia (n=2251) – Maximum F/U = 7.1 yrs – Conversion to AL/MDS = *2.1% 2. Polycythemia vera – Maximum F/U = 7.0 yrs – Conversion to AL/MDS = *2.8% 3. Patients treated with anagrelide for >3 yrs; 0% of ET and 0.26% of P Vera patients transformed *All received prior cytoreductive therapy SM Fruchtman

Anagrelide – Physicians’ Considerations

Why do I need to add anagrelide to my therapeutic armamentarium?

What is unique about anagrelide?

How effective is anagrelide?

What is anagrelide’s safety & toxicity profile?

What factors affect decision to treat?

What outcome can I expect?

HS Gilbert

Evidence-Based Clinical Data in ET

• Comparative phase III clinical trials for rare diseases such as ET are difficult to carry out • No head-to-head data in ET to compare therapeutic agents (hydroxyurea, anagrelide, IFN  , etc) – Shire US initiated head-to-head trial. Discontinued due to inadequate recruitment in HU arm CM Kessler

Pairing MPD Phenotype and Therapy

MyM WBC RBC PLTS

ET

Anagrelide

HS Gilbert

Combined Agent Therapy in MPDs

Treatment of ET: ANAG + IFN-

120 ANAG 100 80 60 40 20 0 IFN-

SPL PLTSX10-4 WBC PCV Weeks

HS Gilbert

Algorithm for Treatment of Thrombocythemia

THROMBOCYTHEMIA Low Risk Age < 40 Age 40-60 Plts 600,000 - 1,000,000/

m

L Plts > 1,000,000/

m

L Age >60 Symptomatic Comorbidities Asymptomatic Other proliferation minimal Other proliferation significant ASA Anagrelide IFN-

Hydroxyurea

HS Gilbert

Treatment: Polycythemia Vera

Objectives of Treatment

• Reduce hematocrit to < 45% • Reduce platelets to < 400,000/ m L • Reduce spleen size • Eliminate monoclonal stem cells • Promote polyclonal hematopoiesis • Prevent myelofibrosis • Avoid leukemogenic therapy HS Gilbert

Risk-Based Treatment for P Vera

• Low risk – Age < 60 years – Platelets < 1,500,000/ m L – No history of thrombosis – No cardiovascular risks • Phlebotomy, target Hct < 45%, and consider low-dose aspirin HS Gilbert

Risk-Based Treatment for P Vera

• Intermediate risk – Age < 60 years – No history of thrombosis – Platelet counts > 1,500,000/ m L or – Cardiovascular risk factors HS Gilbert

Risk-Based Treatment for P Vera

• Intermediate-risk treatment – Phlebotomize to Hct <45% – Treat cardiovascular risk factors – Avoid aspirin if platelet counts > 1,500,000/ m L – Consider IFN  , hydroxyurea, or anagrelide HS Gilbert

Risk-Based Treatment for P Vera

• High risk – Age  60 years or – History of thrombosis • Phlebotomy and hydroxyurea; consider low-dose aspirin, IFN  , anagrelide HS Gilbert

Risk-Based Treatment for P Vera

• High risk – Age > 70 years – Phlebotomy and hydroxyurea and consider low-dose aspirin, radioactive phosphorous; busulfan can be considered HS Gilbert

Women of Childbearing Age with P Vera

• Phlebotomy, target Hct < 45% (42%) • Low-dose aspirin optional - avoid if platelets > 1,500,000/ m L • For high-risk patients, phlebotomy and IFN  HS Gilbert

Phlebotomize to Normal Hct Rise in Hct to 55% Within 1 Year or 10% Rise in 3 Months Randomization Phlebotomy Prn to maintain Hct <45% 32 P 2.7 mc/m 2 IV Q 12 wks prn (limit 5 mc/dose) Phlebotomy for Hct >45% Chlorambucil 10 mg po qd x 6 wks; then qd alternate months Adjust dose for response Phlebotomy for Hct >45%

SM Fruchtman

Normal and Low Cerebral-Blood-Flow Measurements in Relation to Hematocrit

Normal CBF 2 Low CBF 3 040 1 2 3 4 045 6 7 8 9 0 50 1 2 3 4 0 55 6 7 8 9 060 1 2 0 63 4 1 1 3 2 5 4 Hematocrit

SM Fruchtman

Relationship of Cerebral Blood Flow to Hematocrit

60 50 40 30 20 10 0.45

0.50

0.55

Hematocrit 0.60

SM Fruchtman

Sequelae of Treatment

• Phlebotomy to reduce Hct is first-line therapy in P Vera. Sequela is iron deficiency that is usually well tolerated. Frequency of phlebotomy usually decreases with time.

• Low-dose ASA is often effective in preventing symptomatic platelet aggregation in thrombocythemia. Sequela is hemorrhage that is usually minor.

HS Gilbert

Some Myelosuppressive Agents Used for P Vera

Agent

Chlorambucil Busulfan 32 P Hydroxyurea

Pro

Easy to administer Easiest to administer Easy to administer Decreases thrombosis when compared to Phl alone

Con

Leukemogenic Greater megakaryocyte toxicity Pulmonary fibrosis Leukemogenic Vanishing expertise ? Long-term safety Rare toxicities (skin) SM Fruchtman

Agent

ASA

Some Pros and Cons of Agents

Hydroxyurea Anagrelide

Pro

Easy to administer Easy to administer Well tolerated Easy to administer

Con

? Efficacy ? Bleeding ? Dose ? Leukemogenic IFN 32 P ?

Intermittent administration Easiest to give (with experience) Fluid retention CHF ? Long-term safety SC route Drop-out rate ? Long-term safety Leukemogenic Vanishing expertise SM Fruchtman

ECLAP Trial (European Collaboration on Low-Dose ASA in PV) Eligibility Consent Randomization Aspirin 100 mg daily Placebo 12 Month: Follow-up Yearly Follow-up

SM Fruchtman

Collaborative Overview of Randomized Trials of Antiplatelet Therapy

1. 75-325 mg daily equivalent to 500-1500 mg daily 2. Risk of major bleeds small with <325 mg daily 3. Aspirin associated with 25% reduction (

p

<10 -5 ) in arterial vascular events in patients at increased risk for occlusive vascular disease 4. Aspirin halves the risk of deep venous thrombosis in surgical patients SM Fruchtman

Hydroxyurea

• Hydroxyurea (HU) with intermittent phlebotomy (PHL) is a highly efficacious treatment strategy in the management of polycythemia vera (P Vera).

• It is the most frequently employed regimen for patients requiring myelosuppression. (ASH Educational Program, 1992) • In addition, HU is being studied for benign hematologic disorders, such as sickle cell disease.

• Thus, it is imperative to establish its long term safety.

SM Fruchtman

Photo

HS Gilbert

IFN-

 In polycythemia vera • Controls erythrocytosis, reduces spleen size, and relieves pruritis in ~76% of patients • Alternative to anagrelide or hydroxyurea in young patients SM Fruchtman

Long-term Effect of IFN-

in MPDs

TREATMENT OF P Vera: IFN IFN 140 120 100 80 60 40 20 0 WEEKS 7 37 86 145 201 267

PCV PLTS X 10-4 WBC SPL HS Gilbert

Photo

HS Gilbert

IFN Therapy of MPDs: Effect on Spleen and Blood Counts

Agnogenic myeloid metaplasia Post polycythemia myeloid metaplasia Active polycythemia with myeloid metaplasia Polycythemia vera 0 -10 -20 -30 -40 -50 -60 % change in spleen size % change in WBC % change in platelets

SM Fruchtman MPD Research Center.

Combined Agent Therapy in P Vera

1400 1200 1000 800 600 400 200 HU AGR IFN-

PHLEB WBC PLTS PCV 0 0 5 10 15 20 25 HU------------- AGRYLIN---------------- IFN--------- WEEKS 30 35 40

HS Gilbert

Pairing MPD Phenotype and Therapy

MyM WBC RBC PLTS

P Vera

Elderly or failed other agents Hydroxyurea

HS Gilbert

Leukemic Risk with Therapies for ET and P Vera

• Hydroxyurea: Risk (0%-5% in ET; 2.1%-10% in P Vera) increases when HU is combined with other agents – 14% risk of leukemic conversion with HU + 32 P, busulfan, or pipobroman in one study of 357 patients followed for median of 8 years • Busulfan: 3% with busulfan alone • Anagrelide: No evidence of leukemogenicity in patients treated for up to 10 years SM Fruchtman

Spent Phase

• Anemia • Splenomegaly • Nucleated RBC • Tear-drop forms SM Fruchtman

SM Fruchtman

Comparative Incidence of Spent Phase by Protocol and Treatment

On-study events, first 795 weeks of study

Protocol

HU-NPT (08) Phlebotomy (01) Wilcoxon X² 1 Logrank X² 1 = 2.253

= 1.137

Patients

51 134

p

=.1334

p

=.2863

All events HU-NPT (08) Phlebotomy (01) Wilcoxon X² 1 Logrank X² 1 = 1.734

= 0.778

51 134

p

=.1879

p

=.3777

PVSG 8/94.

Total # events

4 15 4 17

%

7.8

11.2

7.8

12.7

SM Fruchtman

Comparative Incidence of Spent Phase/Acute Leukemia by Protocol and Treatment

On-study events, first 795 weeks of study

Protocol

HU-NPT (08) Phlebotomy (01) Wilcoxon X² 1 Logrank X² 1 = 0.119

= 0.045

Patients

51 134

p

=.7301

p

=.8329

Total # events

7 16*

%

13.7

11.9

All events HU-NPT (08) Phlebotomy (01) Wilcoxon X² 1 Logrank X² 1 = 0.196

= 0.235

51 134

p

=.6581

p

=.6277

9 19** 17.6

14.2

*One case has both spent phase and acute leukemia.

**Three cases have both spent phase and acute leukemia.

PVSG 8/94.

SM Fruchtman

Medical Management of Spent Phase P Vera

1.

2.

Transfusion support Others: – Androgens – Low-dose splenic R.T.

– Interferons – anagrelide – erythropoietin – hydroxyurea – research protocols (thalidomide, CSA transplantation) 3.

Splenectomy SM Fruchtman

Treatment: Idiopathic Myelofibrosis

(Myelofibrosis with Myeloid Metaplasia; Agnogenic Myeloid Metaplasia)

Underlying Propensity to Develop Myelofibrosis

• Myelofibrosis is a reactive phenomenon to the abnormality of the pluripotent hematopoietic stem cell. • The fibroblast is not part of the malignant clone. • This in contrast to acute malignant myelofibrosis.

• The fibroblast may be part of the malignant clone.

• Seen in patients with acute leukemic condition.

HS Gilbert

Medical Management of IMF

• Transfusional support • Others: – Androgens – Hydroxyurea – Low-dose splenic RT – Erythropoietin – Interferons – Anagrelide • Splenectomy SM Fruchtman

Splenectomy in IMF

(Tefferi ASH 1998 vs Barosi, Blood 91:3630,1998)

• Mayo data (223 cases) – Median age, 65 years – Mortality, 9%; morb., 31% – Symptoms improved in >90% – Anemia improved in 30% – Thrombocytopenia in 25% – Leukemia in 16% at 2-yr follow-up – Hepatomegaly in 16% – Thrombocytosis in 20% – Median postop survival 2 yrs – Platelet count <50k is risky • Italian multicenter (87 cases) – Compared to 462 non splenectomized cases – Leukemia—26% vs 12% – Leukemia at 12 years after diagnosis —55% vs 27% – Disease presentation with blasts or low platelets is a risk factor for leukemia – HU therapy was not a risk factor – PPMM and PTMM excluded SM Fruchtman

Interference with Collagen Metabolism

Target

Hydroxylation Secretion

Mechanism

Substrate modification Microtubules Microfilaments

Agent

Hydroxyproline analogues Colchicine Vinblastine Cytochalasin B Polymerization Complexing chains Cleaving cross links Lysyl oxidase inhibition D-Penicillamine Potaba p-aminoproprionitrile Degradation Increase collagenase Colchicine SM Fruchtman

We NEED Antifibrosing Therapies

• Marrow • Lung • Liver • Skin • Penis SM Fruchtman

Acute Leukemia and IMF in PVSG-01

Treatment

Phlebotomy Chlorambucil 32 P Total

MM/MF

No Yes No Yes No Yes No Yes

Cases

120 14 127 14 140 16 387 44

Acute Leukemia Cases

1 1 14 5 12 4 27 10

Percent

0.8

7.1

11.0

36.0

8.6

25.0

7.0

23.0

SM Fruchtman

Effect of IFN-

on Splenomegaly

40 35 30 25 20 15 10 5 0 PRE Rx POST Rx 1 2 3 4 POST- P Vera IMF 5 6

HS Gilbert

Effect of IFN-

on Splenomegaly

40 35 30 25 20 15 10 5 0 1 2 3 4 5 6 7 8 IMF 9 10 11 12 13 14 PRE Rx POST Rx

HS Gilbert

Effect of Therapy on Control of Proliferation

MyM WBC RBC PLTS Anagrelide IFN Hydroxyurea

HS Gilbert

Pairing MPD Phenotype and Therapy

MyM

MyM WBC RBC PLTS

P Vera

IFN

HS Gilbert

Combined Agent Therapy in MPDs

MyM

IFN Anagrelide Hydroxyurea

P Vera ET

HS Gilbert

Three Classes of Drugs Used for Treatment of Thrombocythemia in MPDs

DRUG ANAGRELIDE HYDROXYUREA IFN α CLASS OF DRUG MECHANISM OF ACTION EFFECT ON CLONAL vs NORMAL PHPC EFFECT ON MEGAKARYOCYTES Prostaglandin synthetase inhibitor Inhibitor of megakaryocyte maturation and platelet budding None; targets megakaryocytes Prevents maturation and budding Ribonucleoside diphosphate reductase inhibitor Biological response modifier DNA synthesis inhibitor; myelosuppressive Myelosuppressive Immunomodulatory Nonselective Selective; reduces and/or changes MPD clone Decreases by inhibiting proliferation of PHPC Decreases by inhibiting proliferation of PHPC

HS Gilbert

Three Classes of Drugs Used for Treatment of Thrombocythemia in MPDs

DRUG ANAGRELIDE HYDROXYUREA IFN α REDUCTION AND MAINTENANCE OF PLATELETS TO <400,000/ µL Achieved by titration; Gradual reduction; Steady maintenance while on drug.

Difficult to achieve; Difficult to maintain; Requires continuous drug for maintenance.

REDUCTION OF SPLENOMEGALY/ MYELOID METAPLASIA None PREVENTION OR REVERSAL OF FIBROSIS Potentially by effect on megakaryocytes.

ACUTE LEUKEMIA OR MDS TRANS FORMATION Unlikely Unknown None None Increased 5%-10% Achieved by titration; Gradual reduction; Steady maintenance that persists after drug stopped.

Spleen shrinks beginning at 2 mos, continues during 6-12 mos, and persists after drug stopped.

Potentially by changing MPD clone and marrow milieu.

Unlikely Unknown

HS Gilbert

Three Classes of Drugs Used for Treatment of Thrombocythemia in MPDs

DRUG ANAGRELIDE HYDROXYUREA IFN α Good STABILITY OF RESPONSE DURING TREATMENT Good Short without rebound DURATION OF EFFECT AFTER STOPPING TREATMENT SIDE EFFECTS USE DURING PREGNANCY COST ACUTE LEUKEMIA/MDS TRANSFORMATION 2-3+ No $5.00/capsule Unknown Unlikely Fair Short with rebound 1-2+ No $1.00/capsule 5%-10% Long without rebound 2-4+ Acceptable $8.00/MU Unknown Unlikely

HS Gilbert

Three Classes of Drugs Used for Treatment of Thrombocythemia in MPDs

DRUG ANAGRELIDE HYDROXYUREA IFN α Good STABILITY OF RESPONSE DURING TREATMENT Good Fair Short without rebound Short with rebound DURATION OF EFFECT AFTER STOPPING TREATMENT SIDE EFFECTS USE DURING PREGNANCY COST ACUTE LEUKEMIA/MDS TRANSFORMATION 2-3+ No $5.00/capsule Unknown Unlikely 1-2+ No $1.00/capsule 5%-10% Long without rebound 2-4+ Acceptable $8.00/MU Unknown Unlikely

HS Gilbert

ANAGRELIDE

Side Effects of Therapy

HYDROXYUREA IFN α Prostaglandin Inhibition Vasodilator: Headache Dizziness Inotrope: “Palpitations” Renal blood flow: Fluid retention Lactose vehicle Bloating, diarrhea Nausea, vomiting CHF, PULMONARY EDEMA Antimetabolite Myelosuppression: Neutropenia Anemia Dermal: Mucositis Hyperpigmentation Ulceration Gastrointestinal: Nausea, vomiting Bloating, diarrhea Liver damage Systemic: Headache, nausea, Asthenia Biologic Response Modifier Systemic: Fever, chills Myalgia, fatigue Anorexia Cytoreductive: Alopecia Diarrhea Myelosuppression: Neutropenia Anemia Neurologic: Headache, depression Neuropathy Immunomodulator: Hypothyroidism LEUKEMOGENIC MUTAGENIC

HS Gilbert

Treatment: Stem Cell Transplant

Experimental

• Therapies for idiopathic myelofibrosis • Allogeneic stem cells • Autologous stem cells SM Fruchtman

Allogeneic Bone Marrow Transplantation

DONOR usually HLA matched sibling c 750 ml bone marrow aspirated from iliac crest High-dose chemotherapy ± total body irradiation bone marrow graft: donor marrow infused intravenously RECIPIENT ± T cell depletion by monoclonal antibodies Intensive support therapy, e.g. red cells & ± platelets, antibiotics attempts to prevent GVHD, e.g. cyclosporin ± methotrexate

SM Fruchtman

The Sheffield Schema for Predicting Survival

Age (yrs)

<68

Hb (g/dL)

 10 >10

Karyotype

N A N A

Median survival

54 22 180 72 >68  10 >10 N A N A 44 16 70 78 N=normal, A=abnormal SM Fruchtman

The LILLE Scoring System for Predicting Survival

No. of adverse prognostic factors

0

Risk group

Low

Cases (%)

47

Median survival (months)

93 1 Intermediate 45 26 2 High 8 Adverse prognostic factors; Hb <10g/dL, WBC <4 or >30 x 10 9 /L 13 SM Fruchtman

Diagnosis

P Vera

Age

44 P Vera 31

Gender

F

Time from DX to transplant (months)

156 F 144 ET ET ET

P Vera, ET, and Allogeneic BMT

18 49 31 F M F 60 37 180

Medical RX

Hydrea, Phl Hydrea, ASA ASA, Persantin Hydrea Epo, Hydrea

Spleen size

Resected Mildly enlarged Mildly enlarged Mildly enlarged Mildly enlarged SM Fruchtman

Hematologic Parameters

Diagnosis

P Vera

WBC*

5.9

Blasts

<1

Platelet*

879

HCT%

38.2

Grade of fibrosis

3

Cytogenetics

Normal P Vera ET 13.8

11.7

<1 1 ET 14.8

ET *x10 9 cells/L 5.5

1 0 966 1,256 40.2

38.4

252 554 38.6

34.0

1 2-3 2 2-3 Normal Normal Normal Normal SM Fruchtman

Transplantation Characteristics and Outcome

Diagnosis

P Vera P Vera

Donor

HLA ID VUD HLA ID SIB

GVHD

GR 1 Acute Chronic extensive GR 2 Acute Chronic extensive ET ET ET HLA ID SIB HLA ID SIB HLA ID SIB None Grade 3 Acute None Prep Regimen BU/Cy GVHD Prophylaxis CSA/MTX

Comments

On immunosuppression Alive (2.1 yrs) 100% Performance On immunosuppression Alive (1.7 yrs) 80% Performance Alive (7.1 yrs) 100% Performance Alive (1.4 yrs) 100% Performance Alive (1.2 yrs) 80% Performance SM Fruchtman

Cost-effectiveness

Treatment Model: ET

• Treatment in ET: based on high-risk patients • Treatment options include Hydroxyurea (HU), anagrelide and interferon alpha.

• Controversy regarding best choice of agent.

CL Bennett

Available Evidence

• Comparative phase III clinical trials for rare diseases such as ET are difficult to carry out • No head to head comparison data are available for choosing best therapeutic agent in ET CL Bennett

Complications of Untreated ET

• Angina/MI 4.3% /year • TIA/Stroke 3.6% /year • GI or other hemorrhage 0.2% /year • Venous/Arterial thrombosis 2.9% /year CL Bennett

Hydroxyurea

• Typical effective dose 1g/day; – Cost: $142/month • Response rate: 80%-89% • Often used as first-line therapy for ET • Concerns: long-term tolerability and leukemogenicity CL Bennett

Anagrelide

• Maintenance Dose 2.5mg daily; • Cost: $520/month • Adverse effects: Palpitations, (10%-27%), tachycardia & other arrhythmias (< 10%), CHF exacerbation (<2%).

• Response rate: 90% CL Bennett

IFN-

 • Typical effective dose 30 MU/week; Cost (including injection supplies) $1860/month • Withdrawal secondary to adverse effects: 25% 1 st year; 25% additional in 2 nd year • Response rate: 80% CL Bennett

Methods

• A Markov model was used to derive life expectancy and lifetime risk of sequelae of ET and treatments.

• Data derived from the Markov model was then used in a decision tree to compare relative benefits and life-time costs of treatment and treatment-related sequelae of HU, anagrelide and IFN  . CL Bennett

Costs of Complications

• Stroke • MI • GI bleed • TIA • Venous thrombosis • Acute Leukemia $30,000 $30,000 $5,000 $4,700 $3,200 $245,000 CL Bennett

Assumptions of Model

• Leukemia risk modeled as part of sensitivity analysis (baseline risk of 10%) • Markov model allows for only one complication in any one cycle.

• It takes three months to evaluate efficacy of drug.

• Treatment of leukemia results in cure of ET (if survive).

CL Bennett

Markov Model

CL Bennett

Estimated Cost/Effectiveness of Cytoreductive Therapy

Drug Cost [C] Effectiveness [E] (years of life) Marginal C/E [$/LYS] HU $78,000 20.677

$93,073 Anagre lide $132,000 IFN $148,000 21.266

21.229

(Dominated) Costs and cost-effectiveness estimates for a 40 year old individual with ET who is treated with HU, anagrelide, or IFN.

CL Bennett

Cost effectiveness Results (Cont’d)

• $75,000 to $100,000 per life year gained is used as threshold for "cost-effective" • For younger high-risk patients, anagrelide is clinically effective and in the range of options that are considered cost-effective • Hydroxyurea is a cost-effective option for older individuals • IFN  is never cost-effective CL Bennett

Limitations

• No reliable estimate of leukemic risk CL Bennett

Conclusion

Anagrelide is a cost-effective option for younger, symptomatic patients, taking into account a risk of leukemic conversion with hydroxyurea.

Hydroxyurea is a cost-effective option for older patients.

IFN  is never cost-effective and should be restricted to situations such as pregnancy CL Bennett

Conclusions

A New Treatment Dynamic?

• Reduce platelets to low normal range • Is IFN  underutilized? Does it affect the clone more than other approaches?

• Is ASA warranted in all cases? Prescreen with platelet aggregation studies?

• Reserve hydroxyurea for refractory cases?

• Begin treatment earlier – Thrombotic complications are not uncommon in young patients. Early, more aggressive treatment may reduce future complications CM Kessler

Conclusion —P Vera, ET

1. Hydroxyurea has become an old friend (? Enemy).

2. It is an excellent choice for older patients and those at risk for thrombosis.

3. No myelosuppressive therapy is an excellent option for young patients and those at low thrombotic risk.

4. Medium-dose aspirin (75-325 mg daily). The risk/benefit ratio remains to be determined.

SM Fruchtman

Modern Therapeutic Options in MPD

Essential Thrombocythemia Hydroxyurea Anagrelide ASA 32 P IFN-

Plateletpheresis

SM Fruchtman

Take-Home Messages

• Diagnose ET earlier – Be more suspicious – Criteria now permit diagnosis at platelet count of 400,000/ m L – Treat ET more aggressively – Normal platelet count should be therapeutic goal • Use low-dose aspirin if platelet count permits – Helps counteract undetected spontaneous aggregation – No increased bleeding risk until platelets > 1.5 million/ m L RM Petitt

Future Directions for MPD

• Anagrelide – Sustained release ?

– Congeners • IFN – Pegylated IFN – now on market – Albumin-bound IFN – coming • Marrow curettage + autologous CD34 infusion – Better results after splenectomy?

• Angiogenesis inhibitors – Thalidomide – Over 40 others in various phases of evaluation • Other biologic response modifiers RM Petitt

References

References

1.

2.

3.

4.

5.

6.

7.

8.

9.

Tefferi A, Silverstein MN, Hoagland HC. Primary thrombocythemia. Sem Onc 1995, 22:334-340. Gilbert HS. Diagnosis and treatment of polycythemia vera, agnogenic myeloid metaplasia, and essential thrombocythemia. In Neoplastic Diseases of the Blood. Wiernik PH, Canellos GP, Kyle RA, Schiffer CA, (eds): New York: Churchill Livingstone, 1991, p 123.

Hachulla E, Rose C, Trillot N, Caulier-Leleu MT, Pasturel-Michon U. [What vascular events suggest a myeloproliferative disorder]?

J Mal Vasc 2000, 25(5):382-387.

Tefferi A. Chronic myeloid disorders: Classification and treatment overview. Semin Hematol 2001, 38 (1 Suppl 2):1-4.

Nosslinger T, Reisner R, Gruner H, Tuchler H, Nowotny H, Pittermann E, Pfeilstocker M. Dysplastic versus proliferative CMML--a retrospective analysis of 91 patients from a single institution. Leuk Res 2001, 25:741-747.

Westbrook CA, Hsu WT, Chyna B, Litvak D, Raza A, Horrigan SK. Cytogenetic and molecular diagnosis of chromosome 5 deletions in myelodysplasia. Br J Haematol 2000, 110:847-855.

Li S, Salhany KE. Spurious elevation of automated platelet counts in secondary acute monocytic leukemia associated with tumor lysis syndrome. Arch Pathol Lab Med 1999, 123:1111-1114.

Mazur EM, Rosmarin AG, Sohl PA, Newton JL, Narendran A. Analysis of the mechanism of anagrelide-induced thrombocytopenia in humans. Blood 1992, 79:1931-1937.

Solberg LA, Tefferi A, Oles KJ, et al. The effects of anagrelide on human megakaryocytopoiesis. Br J Haematol 1997, 99:174-180.

10.

11.

12.

13.

14.

15.

16.

17.

18.

Tomer A. Effects of anagrelide on in vivo megakaryocyte proliferation and maturation in essential thrombocythemia. Blood 2002, 99:1602-1609.

Murphy S, Peterson P, Iland H, Laslo J. Experience of the Polycythemia Vera Study Group with essential thrombocythemia: a final report on the diagnostic criteria, survival and leukemic transition by treatment. Semin Hematol 1997, 34:29-39.

Cortelazzo S, Finazzi G, Ruggeri M, et al. Hydroxyurea for patients with essential thrombocythemia and a high risk of thrombosis.

N Engl J Med 1995, 332:1132-1136.

Ruggeri M, Finazzi G, Tosetto A, Riva S, Rodeghiero F, Barbui T. No treatment for low-risk thrombocythaemia: results from a prospective study. Br J Haematol 1998, 103:772-777.

Michiels JJ. Aspirin and platelet-lowering agents for the prevention of vascular complications in essential thrombocythemia. Clin Appl Thromb Hemost 1999, 5:247-251.

Jensen MK, de Nully Brown P, Nielsen OJ, Hasselbalch HC. Incidence, clinical features and outcome of essential thrombocythaemia in a well defined geographical area.

Eur J Haematol 2000, 65(2):132-139.

Mesa RA, Silverstein MN, Jacobsen SJ, Wollan PC, Tefferi A. Population-based incidence and survival figures in essential thrombocythemia and agnogenic myeloid metaplasia: an Olmsted County Study, 1976-1995. Am J Hematol 1999, 61(1):10-15.

Gugliotta L, et al. Poster 289, ISH Stockholm, September 1997.

Georgii A, Buhr T, Buesche G, Kreft A, Choritz H. Classification and staging of Ph-negative myeloproliferative disorders by histopath ology from bone marrow biopsies. Leuk Lymphoma 1996, 22:15-29.

References (Cont’d)

19.

20.

21.

22.

23.

24.

25.

26.

27.

Berk PD, Goldberg JD, Donovan PB, Fruchtman SM, Berlin NI, Wasserman LR. Therapeutic recommendations in polycythemia vera based on Polycythemia Vera Study Group protocols. Semin Hematol 1986, 23(2):132-143.

Storen EC, Tefferi A. Long-term use of anagrelide in young patients with essential thrombocythemia. Blood 2001, 97(4):863-866.

Murphy S. Diagnostic criteria and prognosis in polycythemia vera and essential thrombocythemia. Semin Hematol 1999, 36(1Suppl 2):9-13.

Regev A, Stark P, Blickstein D, Lahav M. Thrombotic complications in essential thrombocythemia with relatively low platelet count.

Am J Hematol 1997, 56:168-172.

Michiels JJ, van Genderen PJ, Lindemans J, van Vliet HH. Erythromelalgic, thrombotic and hemorrhagic manifestations in 50 cases of thrombocythemia. Leuk Lymphoma 1996, 22 (Suppl 1):47-56.

Cortelazzo S, Viero P, Finazzi G, D'Emilio A, Rodeghiero F, Barbui T. Incidence and risk factors for thrombotic complications in a historical cohort of 100 patients with essential thrombocythemia. J Clin Oncol 1990, 8:556-562.

Kurzrock R, Cohen PR. Erythromelalgia and myeloproliferative disorders. Arch Int Med 1989, 149:105-109.

McKusick MA. Imaging findings in Budd-Chiari syndrome. Liver Transpl 2001, 7(8):743-744.

Vandenbroucke JP, Rosing J, Bloemenkamp KW, Middeldorp S, Helmerhorst FM, Bouma BN, Rosendaal FR. Oral contraceptives and the risk of venous thrombosis. N Engl J Med 2001, 344:1527-1535.

28.

29.

30.

31.

32.

33.

34.

35.

36.

Kutti J, Ridell B. Epidemiology of the myeloproliferative disorders: essential thrombocythaemia, polycythaemia vera and idiopathic myelofibrosis. Pathol Biol (Paris) 2001, 49(2):164-166.

Thiele J, Kvasnicka HM. Clinicopathology and histochemistry on bone marrow biopsies in chronic myeloproliferative disorders--a clue to diagnosis and classification. Pathol Biol (Paris) 2001, 49(2):140-147.

Ellis JT, Peterson P, Geller SA, Rappaport H. Studies of the bone marrow in polycythemia vera and the evolution of myelofibrosis and second hematologic malignancies. Semin Hematol 1986, 23(2):144-155.

Rao GHR, ed. Handbook of Platelet Physiology and Pharmacology. Boston: Kluwer Academic, 1999.

Coller BS. Blockade of platelet GPIIb/IIIa receptors as an antithrombotic strategy. Circulation 1995, 92(9):2373-2380.

Boyles J, Fox JE, Phillips DR, Stenberg PE. Organization of the cytoskeleton in resting, discoid platelets: preservation of actin filaments by a modified fixation that prevents osmium damage.

J Cell Biol 1985, 101(4):1463-1472.

Loftus JC, Choate J, Albrecht RM. Platelet activation and cytoskeletal reorganization: high voltage electron microscopic examination of intact and Triton-extracted whole mounts.

J Cell Biol 1984, 98(6):2019-2025.

Bevers EM, Tilly RH, Senden JM, Comfurius P, Zwaal RF. Exposure of endogenous phosphatidylserine at the outer surface of stimulated platelets is reversed by restoration of aminophospholipid translocase activity. Biochemistry 1989, 28(6):2382-2387.

Bennett JS. Novel platelet inhibitors. Annu Rev Med 2001, 52:161-184.

References (Cont’d)

37.

38.

39.

40.

41.

42.

43.

44.

45.

Kulkarni S, Dopheide SM, Yap CL, et al. A revised model of platelet aggregation. J Clin Invest 2000, 105(6):783-791.

Luchtman-Jones L, Broze GJ Jr. The current status of coagulation. Ann Med 1995, 27(1):47-52.

Wehmeier A, Sudhoff T, Meierkord F. Relation of platelet abnormalities to thrombosis and hemorrhage in chronic myeloproliferative disorders. Semin Thromb Hemost 1997, 23:391-402.

Jensen MK, de Nully Brown P, Lund BV, Nielsen OJ, Hasselbalch HC. Increased platelet activation and abnormal membrane glycoprotein content and redistribution in myeloproliferative disorders. Br J Haematol 2000,110:116-124.

Murphy S. Therapeutic dilemmas: balancing the risks of bleeding, thrombosis, and leukemic transformation in myeloproliferative disorders (MPD). Thromb Haemost 1997, 78(1):622-626.

Faurschou M, Nielsen OJ, Jensen MK, Hasselbalch HC. High prevalence of hyperhomocysteinemia due to marginal deficiency of cobalamin or folate in chronic myeloproliferative disorders.

Am J Hematol 2000, 65:136-140.

Horikawa Y, Matsumura I, Hashimoto K, et al. Markedly reduced expression of platelet c-mpl receptor in essential thrombocythemia. Blood 1997, 90(10):4031-4038.

Moliterno AR, Hankins WD, Spivak JL. Impaired expression of the thrombopoietin receptor by platelets from patients with polycythemia vera. N Engl J Med 1998, 338(9):572-580.

Le Blanc K, Andersson P, Samuelsson J. Marked heterogeneity in protein levels and functional integrity of the thrombopoietin receptor c-mpl in polycythaemia vera. Br J Haematol 2000, 108(1):80-85.

46.

47.

48.

49.

50.

51.

52.

53.

54 .

Matsumura I, Horikawa Y, Kanakura Y. Functional roles of thrombopoietin-c-mpl system in essential thrombocythemia.

Leuk Lymphoma 1999, 32(3-4):351-358.

Cardier JE, Foster DC, Lok S, Jacobsen SE, Murphy MJ Jr. Megakaryocytopoiesis in vitro: from the stem cells’ perspective. Stem Cells 1996, 14(Suppl 1):163-172.

McCarty JM, Simanis JP, Kanamori D, Dessypris EN. Anagrelide exhibits antiproliferative effects on megakaryocyte progenitors generated from human CD34+ cells by inhibiting TPO specific, but not IL-3 specific signaling events. Blood 1999, 94(11) 1part2: 1178a.

McCarty JM, Melone PM, Heisey C, Dessypris EN. Differences in the species-specific activity of anagrelide is mediated through the c mpl receptor. Blood 2000, 96(11) 1part2: 3213a.

Tefferi A, Solberg LA, Silverstein MN. A clinical update in polycythemia vera and essential thrombocythemia. Am J Med 2000, 109(2):141-149.

Tefferi A, Fonseca R, Pereira DL, Hoagland HC. A long-term retrospective study of young women with essential thrombocythemia. Mayo Clin Proc 2001, 76(1):22-28.

Berk PD, Goldberg JD, Silverstein MN, et al. Increased incidence of acute leukemia in polycythemia vera associated with chlorambucil therapy. N Engl J Med 1981, 304(8):441-447.

Marchioli R, Landolfi R, Barbui T, Tognoni G. Feasibility of randomised clinical trials in rare diseases: the case of polycythemia vera. Leuk Lymphoma 1996, 22 (Suppl 1):121-127.

Murphy S, Rosenthal DS, Weinfeld A, et al. Essential thrombocythemia: response during first year of therapy with melphalan and radioactive phosphorus: a Polycythemia Vera Study Group report. Cancer Treat Rep 1982, 66(7):1495-1500.

References (Cont’d)

55.

56.

57.

58.

59.

60.

61.

62.

63.

64.

Laszlo J. Myeloproliferative disorders (MPD): myelofibrosis, myelosclerosis, extramedullary hematopoiesis, undifferentiated MPD, and hemorrhagic thrombocythemia. Semin Hematol 1975, 12:409-432.

Iland HJ, Laszlo J, Peterson P, et al. Essential thrombocythemia: clinical and laboratory characteristics at presentation. Trans Assoc Am Physicians 1983, 96:165-174.

Kutti J, Wadenvik H. Diagnostic and differential criteria of essential thrombocythemia and reactive thrombocytosis. Leuk Lymph 1996, 22 (Suppl 1):41-45.

Pearson TC. Evaluation of diagnostic criteria in polycythemia vera. Semin Hematol 2001, 38(1 Suppl 2):21-24.

Pearson TC, Messinezy M. The diagnostic criteria of polycythaemia rubra vera. Leukemia Lymph 1996, 22:87-93.

Pearson, TC. Diagnosis and classification of erythrocytoses and thrombocytoses. Baillieres Clin Haematol 1998, 11:695-720. Michiels JJ, Kutti J, Stark P, et al. Diagnosis, pathogenesis and treatment of the myeloproliferative disorders: essential thrombocythemia, polycythemia vera and essential megakaryocytic granulocytic metaplasia and myelofibrosis.

Neth J Med 1999, 54:46-62.

Vardiman JW, Harris NL, Brunning RD. The World Health Organization (WHO) classification of the myeloid neoplasms. Blood 2002, 100:2292-2302.

Chievitz E, Thiede T. Complications and causes of death in polycythemia vera. Acta Medica Scandinavica 1962, 172:513-523.

Fruchtman SM, Mack K, Kaplan ME, Peterson P, Berk PD, Wasserman LR. From efficacy to safety: a Polycythemia Vera Study Group report on hydroxyurea in patients with polycythemia vera. Semin Hematol 1997, 34(1):17-23.

65.

66.

67.

68.

69.

70.

71.

72.

73.

Nand S, Messmore H, Fisher SG, Bird ML, Schulz W, Fisher RI. Leukemic transformation in polycythemia vera: analysis of risk factors. Am J Hematol 1990, 34(1):32-36.

Nand S, Stock W, Godwin J, Fisher SG. Leukemogenic risk of hydroxyurea therapy in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis.

Am J Hematol 1996, 52(1):42-46.

Randi ML, Rossi C, Fabris F, Menapace L, Girolami A. Aspirin seems as effective as myelosuppressive agents in the prevention of rethrombosis in essential thrombocythemia. Clin Appl Thromb Hemost 1999, 5(2):131-135.

Dewald GW, Wright PI. Chromosome abnormalities in the myeloproliferative disorders. Semin Oncol 1995, 22(4):341-354.

Tefferi A, Silverstein MN. Treatment of polycythaemia vera and essential thrombocythaemia. Baillieres Clin Haematol 1998, 11(4):769-785.

Jantunen R, Juvonen E, Ikkala E, et al. The predictive value of vascular risk factors and gender for the development of thrombotic complications in essential thrombocythemia.

Ann Hematol 2001, 80:74-78.

Gilbert HS. Diagnosis and treatment of thrombocythemia in myeloproliferative disorders. Oncology (Huntingt) 2001, 5(8):989 996, 998; discussion 999-1000,1006,1008.

Harrison CN, Gale RE, Machin SJ, Linch DC. A large proportion of patients with a diagnosis of essential thrombocythemia do not have a clonal disorder and may be at lower risk of thrombotic complications. Blood 1999, 93(2):417-424.

Tefferi A. Recent progress in the pathogenesis and management of essential thrombocythemia. Leuk Res 2001, 25(5):369-377.

References (Cont’d)

74.

75.

76.

77.

78.

79.

80.

81.

82.

83.

Briere J, Guilmin F. Management of patients with essential thrombocythemia: current concepts and perspectives. Pathol Biol (Paris) 2001, 49:178-183.

Kessler CM, Klein HG, Havlik RJ. Uncontrolled thrombocytosis in chronic myeloproliferative disorders. Br J Haematol 1982, 50(1):157-167.

Berrebi A, Shvidel L, Shtalrid M, Klepfish A. Short course of busulphan in essential thrombocythaemia: remodelling of an old strategy. Br J Haematol 2000, 109:249-250.

Gugliotta L, et al. Abstract 883, EHA Paris, June 1996.

Kiladjian JJ, et al. Abstract 887, EHA Paris, June 1996.

Finazzi G, Ruggeri M, Rodeghiero F, Barbui T. Second malignancies in patients with essential thrombocythaemia treated with busulphan and hydroxyurea: long-term follow-up of a randomized clinical trial. Br J Haematol 2000, 110:577-583.

Gilbert, HS. Historical perspective on the treatment of essential thrombocythemia and polycythemia vera. Semin Hematol 1999, 36(1 Suppl 2):19-22.

Chaine B, Neonato MG, Girot R, Aractingi S. Cutaneous adverse reactions to hydroxyurea in patients with sickle cell disease. Arch Dermatol 2001, 137:467-470.

Weinfeld A, Swolin B, Westin J, et al. Acute leukemia after hydroxyurea treatment in polycythemia vera and allied disorders: Prospective study of efficacy and leukemogenicity with therapeutic implications. Eur J Haematol 1994, 52:134-139.

Randi ML, Stocco F, Rossi C, Tison T, Girolami A. Thrombosis and hemorrhage in thrombocytosis: evaluation of a large cohort of patients (357 cases). J Med 1991, 22(4-5):213-223.

84.

85.

86.

87.

88.

89.

90.

91.

92.

Sterkers Y, Preudhomme C, Lai JL, Demory JL, Caulier MT, Wattel E, Bordessoule D, Bauters F, Fenaux P. Acute myeloid leukemia and myelodysplastic syndromes following essential thrombocythemia treated with hydroxyurea: high proportion of cases with 17p deletion. Blood 1998, 91(2):616-622.

Hanft VN, Fruchtman SR, Pickens CV, Rosse WF, Howard TA, Ware RE. Acquired DNA mutations associated with in vivo hydroxyurea exposure. Blood 2000, 95(11):3589-3593.

Van Genderen PJ, Michiels JJ. Hydroxyurea in essential thrombocytosis. N Engl J Med 1995, 333:802-803.

Lengfelder E, Griesshammer M, Hehlmann R. Interferon-alpha in the treatment of essential thrombocythemia. Leuk Lymphoma 1996, 22 (Suppl 1):135-142.

Lazzarino M, Vitale A, Morra E, et al. Therapy of essential thrombocythemia with alpha-interferon: results and prospects.

Eur J Haematol Suppl 1990, 52:15-21.

Lazzarino M, Vitale A, Morra E, et al. Interferon alpha-2b as treatment for Philadelphia-negative chronic myeloproliferative disorders with excessive thrombocytosis. Br J Haematol 1989, 72:173-177.

Wright CA, Tefferi A. A single institutional experience with 43 pregnancies in essential thrombocythemia. Eur J Haematol 2001, 66(3):152-159.

Brooks WG, Stanley DD, Goode JV. Role of anagrelide in the treatment of thrombocytosis. Ann Pharmacother 1999, 33(10):1116-1121.

Pescatore SL, Lindley C. Anagrelide: a novel agent for the treatment of myeloproliferative disorders. Expert Opin Pharmacother 2000, 1:537-546.

References (Cont’d)

93.

94.

95.

96.

Knutsen H, Hysing J. Anagrelide in primary thrombocythemia. Tidsskr Nor Laegeforen 2001, 121:1478-1482.

No authors listed. Anagrelide, a therapy for thrombocythemic states: experience in 577 patients. Anagrelide Study Group. Am J Med 1992, 92:69-76.

Laguna MS, Kornblihtt LI, Marta RF, Michiels JJ, Molinas FC. Effectiveness of anagrelide in the treatment of symptomatic patients with essential thrombocythemia. Clin Appl Thromb Hemost 2000, 6(3):157-161.

Finazzi G, Barbui T. Treatment of essential thrombocythemia with special emphasis on leukemogenic risk. Ann Hematol 1999, 78(9):389-392.

97.

98.

Tefferi A, Silverstein MN, Petitt RM, Mesa RA, Solberg LA Jr. Anagrelide as a new platelet-lowering agent in essential thrombocythemia: mechanism of action, efficacy, toxicity, current indications. Semin Thromb Hemost 1997, 23(4):379-383.

Silverstein MN, Petrone ME, Petitt RM, Dement MP, Vukovich RA. The safety profile of anagrelide for treatment of thrombocythemia. Blood 1996, 88(Suppl 1):583a (abstr).

99.

Gilbert HS. Other secondary sequelae of treatments for myelo proliferative disorders. Semin Oncol 2002, 29 (3 Suppl 10):22-27.

100. Package insert, anagrelide hydrochloride, Shire US Inc.

101. From unpublished data.

102. Shire Study Report No 13970-301. An open protocol for the use of Agrylin (anagrelide HCl) for patients with thrombocythemia. 2002.

103. Fruchtman SM, Petitt RM, Gilbert H, Fiddler G, Lyne A. Anagrelide Study Group. Blood 2002, 100:256a.

104. Gilbert HS. Current management in polycythemia vera. Semin Hematol 2001, 38(1Suppl 2):25-28.

105. Elliott MA, Tefferi A. Interferon-alpha therapy in polycythemia vera and essential thrombocythemia. Semin Thromb Hemost 1997, 23(5):463-472.

106. Lahtinen R, Kuikka J. Cerebral blood flow in polycythaemia vera. Ann Clin Res 1983, 15(5-6):200-202.

107. Michiels JJ, Barbui T, Finazzi G, Fuchtman SM, Kutti J, Rain JD, Silver RT, Tefferi A, Thiele J. Diagnosis and treatment of polycythemia vera and possible future study designs of the PVSG. Leuk Lymphoma 2000, 36(3-4):239-253.

108. Landolfi R, Marchioli R. European Collaboration on Low-dose Aspirin in Polycythemia Vera (ECLAP): a randomized trial. Semin Thromb Hemost 1997, 23(5):473-478.

109. No authors listed. Collaborative overview of randomised trials of antiplatelet therapy--III: Reduction in venous thrombosis and pulmonary embolism by antiplatelet prophylaxis among surgical and medical patients. Antiplatelet Trialists' Collaboration. BMJ 1994, 308(6923):235-246.

110. No authors listed. Collaborative overview of randomised trials of antiplatelet therapy--II: Maintenance of vascular graft or arterial patency by antiplatelet therapy. Antiplatelet Trialists' Collaboration. BMJ 1994, 308(6922):159-168. Review.

111. No authors listed. Collaborative overview of randomised trials of antiplatelet therapy--I: Prevention of death, myocardial infarction, and stroke by prolonged antiplatelet therapy in various categories of patients. Antiplatelet Trialists’ Collaboration. BMJ 1994, 308(6921):81-106.

112. Lengfelder E, Berger U, Hehlmann R. Interferon alpha in the treatment of polycythemia vera. Ann Hematol 2000, 79(3):103-109.

References (Cont’d)

113. Gilbert HS. Long term treatment of myeloproliferative disease with interferon-alpha-2b: feasibility and efficacy. Cancer 1998, 83(6):1205-1213.

114. Butcher C, D'Andrea RJ. Molecular aspects of polycythemia vera (review). Int J Mol Med 2000, 6(3):243-252.

115. Thiele J, Kvasnicka HM, Zankovich R, Diehl V. Relevance of bone marrow features in the differential diagnosis between essential thrombocythemia and early stage idiopathic myelofibrosis. Haematologica 2000, 85(11):1126-1134.

116. Tefferi A, Mesa RA, Nagorney DM, Schroeder G, Silverstein MN. Splenectomy in myelofibrosis with myeloid metaplasia: a single institution experience with 223 patients. Blood 2000, 95(7):2226 2233.

117. Barosi G, Ambrosetti A, Centra A, et al. Splenectomy and risk of blast transformation in myelofibrosis with myeloid metaplasia. Italian Cooperative Study Group on Myeloid with Myeloid Metaplasia. Blood 1998, 91(10):3630-3636.

118. Myllyharju J, Kivirikko KI. Collagens and collagen-related diseases. Ann Med 2001, 33(1):7-21.

119. Sacchi S. The role of alpha-interferon in essential thrombocythaemia, polycythaemia vera and myelofibrosis with myeloid metaplasia (MMM): a concise update. Leuk Lymphoma 1995,19:13-20.

120. Gilbert HS. The role of anagrelide, hydroxyurea, and interferon Education Program Book, 1999, pp 141-143.

α in treating the thrombocythemia of myeloproliferative disease: a new approach for the millennium. International Society of Hematology 121. Bennett CL, Weinberg PO, Golub RM. Cost-effectiveness model of a phase II clinical trial of a new pharmaceutical for essential thrombocythemia: is it helpful to policy makers? Semin Hematol 1999, 36(1Suppl 2):26-29.

122. Lubbert M, Wijermans P, Kunzmann R, et al. Cytogenetic responses in high-risk myelodysplastic syndrome following low dose treatment with the DNA methylation inhibitor 5-aza-2' deoxycytidine. Br J Haematol 2001, 114(2):349-357.

123. Shimamoto T, Iguchi T, Ando K, et al. Successful treatment with cyclosporin A for myelodysplastic syndrome with erythroid hypoplasia associated with T-cell receptor gene rearrangements.

Br J Haematol 2001, 114(2):358-361.

124. Reilly JT, Snowden JA, Spearing RL, Fitzgerald PM, Jones N, Witmore A, Potter A. Cytogenetic abnormalities and their prognostic significance in idiopathic myelofibrosis: a study of 106 cases.

Br J Haematol 1997, 98:96-102.

125. Jurado M, Deeg H, Gooley T, et al. Haemopoietic stem cell transplantation for advanced polycythaemia vera or essential thrombocythaemia. Br J Haematol 2001, 112(2):392-396.

126. Dupriez B, Morel P, Demory JL, Lai JL, Simon M, Plantier I, Bauters F. Prognostic factors in agnogenic myeloid metaplasia: a report on 195 cases with a new scoring system. Blood 1996, 88:1013-1018.

127. Anderson JE, Sale G, Appelbaum FR, Chauncey TR, Storb R. Allogeneic marrow transplantation for primary myelofibrosis and myelofibrosis secondary to polycythaemia vera or essential thrombocytosis. Br J Haematol 1997, 98(4):1010-1016.

References (Cont’d)

128. Barbui T, Finazzi G, Dupuy E, Kiladjian JJ, Briere J. Treatment strategies in essential thrombocythemia. A critical appraisal of various experiences in different centers. Leuk Lymphoma 1996, 22 (Suppl 1):149-160.

129. Silverstein MN, Petitt RM, Solberg LA Jr, Fleming JS, Knight RC, Schacter LP. Anagrelide: a new drug for treating thrombocytosis.

N Engl J Med 1988, 318:1292-1294.

130. Sacchi S, Tabilio A, Leoni P, Riccardi A, Vecchi A, Messora C, Falzetti F, Rupoli S, Ucci G, Martelli MF. Interferon alpha-2b in the long-term treatment of essential thrombocythemia.

Ann Hematol 1991, 63:206-209.

131. Northwestern Memorial Hospital Pharmacy (for costs, dc) 132. Golub R, Adams J, Dave S, Bennett CL. Cost-effectiveness considerations in the treatment of essential thrombocythemia. Semin Oncol 2002, 29(3 Suppl 10):28-32.

133. Holloway RG, Witter DM Jr, Lawton KB, Lipscomb J, Samsa G. Inpatient costs of specific cerebrovascular events at five academic medical centers. Neurology 1996, 46:854-860.

134. Waters TM, Bennett CL, Pajeau TS, Sobocinski KA, Klein JP, Rowlings PA, Horowitz MM. Economic analyses of bone marrow and blood stem cell transplantation for leukemias and lymphoma: what do we know? Bone Marrow Transplant 1998, 21:641-650.

This ends the Non-CML MPD Slide Kit CD-ROM program. If you would like to review a slide, left click the HOUSE icon, which will return you to the Table of Contents, and proceed from there. Otherwise, left click EXIT to end program.

Explanatory notes and references accompany each slide and are located in the binder, along with the CME posttest and evaluation.

We hope you find this material useful in your practice.

Exit