Aplastic Anemia: Current Thinking LGL on the Disease ... AA Current... · aplastic anemia = low...

14
3/23/2015 1 Aplastic Anemia: Current Thinking on the Disease, Diagnosis, and Non-Transplant Treatment Bogdan Dumitriu, MD Hematology Branch National, Heart, Lung and Blood Institute National Institutes of Health BONE MARROW FAILURE SYNDROMES SDS DKC LGL AA AA/PNH PNH MDS hypocellular MDS AML AID: MS, IBD, uveitis, DM type 1, etc. Hemoglobin variable, around 7-8 g/dL (>13 g/dL) weakness, fatigue, shortness of breath, lack of energy Neutrophils under 500/uL (>1500/uL) infections, mouth ulcers Platelets under 10.000/uL (>150.000/uL) bleeding (gums, nose, skin) APLASTIC ANEMIA = LOW BLOOD COUNTS CLINICAL MANIFESTATIONS OF BONE MAROW FAILURE anemia, bleeding, infection AGE AT DIAGNOSIS Aplastic Anemia Admissions to NIH Clinical Center Years Camitta et al, Blood 53:504, 1979 Williams et al, Sem Hematol 10:195, 1973 6 5 4 3 2 1 0 0 60 80 100 20 40 Utah, extrapolated severe NATURAL HISTORYOF APLASTIC ANEMIA % Surviving AA Study Group, non-transplanted (n = 63) Utah, total (n = 99) Severity Criteria (two of three): platelets <20k/uL reticulocytes <1% (60k/uL) ANC <500/uL Very severe: ANC <200/uL

Transcript of Aplastic Anemia: Current Thinking LGL on the Disease ... AA Current... · aplastic anemia = low...

3/23/2015

1

Aplastic Anemia: Current Thinking

on the Disease, Diagnosis, and

Non-Transplant Treatment

Bogdan Dumitriu, MD

Hematology Branch

National, Heart, Lung and Blood Institute

National Institutes of Health

BONE MARROW FAILURE SYNDROMES

SDS

DKC

LGL

AA

AA/PNH

PNH

MDShypocellular

MDS

AML

AID: MS, IBD,

uveitis, DM

type 1, etc.

Hemoglobin – variable, around 7-8 g/dL (>13 g/dL)

weakness, fatigue, shortness of breath, lack of energy

Neutrophils – under 500/uL (>1500/uL)

infections, mouth ulcers

Platelets – under 10.000/uL (>150.000/uL)

bleeding (gums, nose, skin)

APLASTIC ANEMIA = LOW BLOOD COUNTS CLINICAL MANIFESTATIONS OF BONE MAROW FAILURE

anemia, bleeding, infection

AGE AT DIAGNOSIS

Aplastic Anemia Admissions to NIH Clinical Center

YearsCamitta et al, Blood 53:504, 1979

Williams et al, Sem Hematol 10:195, 1973

65432100

60

80

100

20

40

Utah, extrapolated severe

“NATURAL HISTORY” OF APLASTIC ANEMIA

% S

urv

ivin

g

AA

Study

Group,

non-transplanted (n = 63)

Utah, total (n = 99)

Severity Criteria (two of three):

platelets <20k/uL

reticulocytes <1% (60k/uL)

ANC <500/uL

Very severe: ANC <200/uL

3/23/2015

2

Normal

bone

marrow

Aplastic

anemiaPathophysiology of Aplastic Anemia

Stem cells

Hematopoietic

progenitors

Immune attack

Circulating

blood

cells

Stem cells

Hematopoietic

progenitors

Circulating

blood

cells

Most of the cases of Aplastic Anemia have no identifiable cause

There are inherited (genetic) causes of aplastic anemia/bone marrow failure

Pregnancy, eosinophilic fasciitis, and seronegative hepatitis are associated with AA

Drugs and chemicals have been reported as well (Benzene, Chloramphenicol)

All identifiable causes explain very few cases of AA

Causes of Aplastic Anemia

• 1960’s 10% survival in 1 year

• 2010 90% survival in 1 year

• Immunosuppressive therapy

• Bone marrow transplantation

• Supportive care

• Novel agents

2

1

3

45

6

SAA treatment algorithm

3/23/2015

3

•Anti-thymocyte globulin (ATG)

• Horse

• Rabbit

• Cyclosporine (CsA)

• Campath

• Others (MMF, sirolimus, tacrolimus, cyclophosphamide)

Immunosuppressive therapy

• First line of treatment (vs. transplant)

• Salvage for treatment-refractory patients

• Treatment for relapsed disease

Immunosuppressive therapy

Cytotoxicity

assay

Immunization with

human thymocytes

Anti-human

thymocytes

antibodies

T

Purification

of seraIgG

ATG

Anti-thymocyte Globulin (ATG) Production

Thymus

RESPONSE OF SEVERE APLASTIC ANEMIA TO

INTENSIVE IMMUNOSUPPRESSION

7,000

6,000

5,000

4,000

3,000

2,000

1,000

024-Sep 4-Oct 14-Oct 24-Oct 3-Nov 13-Nov 23-Nov 3-Dec 13-Dec 23-Dec 2-Jan 12-Jan 22-Jan

300

250

200

150

100

50

024-Sep 4-Oct 14-Oct 24-Oct 3-Nov 13-Nov 23-Nov 3-Dec 13-Dec 23-Dec 2-Jan 12-Jan 22-Jan

TxTx Tx

Tx

CSA

ATG

42

40

38

34

32

30

26

2424-Sep 14-Oct 3-Nov 23-Nov 13-Dec 2-Jan 22-Jan

36

28

10,000

15,000

20,000

25,000

30,000

35,000

40,000

45,000

50,000

55,000

Tx Tx

ANC

Platelets

ReticHct

Immunosuppressive therapy

at diagnosis of SAA

ATG AND CSA FOR SEVERE APLASTIC ANEMIA

OVERALL SURVIVAL

1.0

0.8

0.6

0.4

0.2

0.00 1000 2000 3000

Days

4000

Su

rviv

al

60% response rate

3/23/2015

4

Study Years NMedian Age

(years)Response

Relaps

e

Clonal

EvolutionSurvival

German 1986-1989 84 32 65% 19% 8% 58% at 11 yrs

NIH 1991-1998 122 35 61% 35% 11% 55% at 7 yrs

EGMBT 1991-1998 100 16 77% 12% 11% 87% at 5 yrs

Japan 1992-1997 119 9 68% 22% 6% 88% at 3 yrs

German/A

ustrian

1993-1997 114 9 77% 12% 6% 87% at 4yrs

Japan 1996-2000 101 54 74% 42% 8% 88% at 4 yrs

NIH 1999-2003 104 30 62% 37% 9% 80% at 4 yrs

EGBMT 2002-2008 192 46 70% 33% 4% 76% at 6 yrs

NIH 2003-2005 77 26 57% 26% 10% 93% at 3yrs

NIH 2005-2010 120 28 68% 28% 21% 96% at 3 yrs

Young NS, Calado RT, Scheinberg P. Blood 2006

INTENSIVE IMMUNOSUPPRESSION FOR SAA

COMPARISON OF RESULTS

• Add to horse ATG + CsA platform

– G-CSF (Neupogen)

– Mycophenolate mofetil

– Sirolimus

– long course immunosuppression

• Augment initial lymphocytotoxicity

– Rabbit ATG

– Campath

NEW DIRECTIONS IN TREATMENT

FOR APLASTIC ANEMIA

A Randomized Trial of H-ATG vs. R-ATG in SAA

Patients and Methods

• 120 consecutive patients (60 per arm)

• NIH Clinical Center

• 1:1 randomization

• Primary objective –response at 6 months

Scheinberg et al. NEJM 2011

Horse ATG Rabbit ATG P-value

3 months 37/60 (62%) 20/60 (33%) 0.003

6 months 41/60 (68%) 22/60 (37%) < 0.001

A Randomized Trial of H-ATG vs. R-ATG in SAA

Hematologic Responses at 3 and 6 months

A Randomized Trial of H-ATG vs. R-ATG in SAA

Blood Count Recovery in Responders

Months

Horse ATG Rabbit ATG

Absolute

reticulocyte

count

Absolute

neutrophil

count

Platelets

630630

120,000

80,000

40,000

0

0

1,000

2,000

100,000

10,000

Num

ber

per L

For new diagnosed SAA

recommended/preferred

immunosuppression regimen

is horse ATG/CSA

3/23/2015

5

Immunosuppressive therapy

for refractory SAA

• rabbit ATG

• Campath

0 250 500 750 10000

25

50

75

100

Time in days

Perc

en

t su

rviv

al

Survival of refractory SAA following retreatment

with rabbit ATG + CsA (salvage)

responders

non-responders

Scheinberg P, Nunez O, Young NS. Br J Haematol 2006

1/3 Response Rate

Alemtuzumab (Campath-1H)

• Anti-CD52 Antibody

• Murine hypervariable regions fused into human IgG1

• CD52 expressed:– B and T cells

– NK cells, dendritic cells

– Monocytes, macrophages

– Plasma cells, Eos

• No CD52 expression on:– RBCs, platelets

– Hematopoietic stem cells

Ravandi and O’Brien, Cancer Invest. 2007 24: 718-725

Hernández-Campo PM, Cytometry B Clin Cytom. 2006 70:71

SECOND IMMUNOSUPPRESSION FOR

REFRACTORY SAA

Treatment arm (N=54) Overall response

rabbit ATG (N=27) 9 (35%)

alemtuzumab (N=27) 10 (37%)

For refractory SAA salvage

immunosuppression with

either rabbit ATG or Campath

rescues 1/3 of patients

Immunosuppressive therapy

for relapsed SAA

• cyclosporine

• rabbit ATG

• alemtuzumab

3/23/2015

6

ATG AND CSA FOR SEVERE APLASTIC ANEMIA

RELAPSE

1.0

0.8

0.6

0.4

0.2

0.0

0 1000 2000 3000

Days

4000

0

Pro

po

rtio

n r

elap

sin

g

RELAPSE AFTER ATG + CSA

Cyclosporine-dependence Post-1strelapse

Years post-relapse 1 2 3 4 5 6 7

Patients on CsA 20/22 19/20 14/18 11/17 11/14 7/11 4/7

(86%) (91) (78) (65) (79) (64) (57)

Retreatment with rabbit ATG + CsA Post-1strelapse 2/3 response

Rosenfeld S, Follmann D, Nunez O, Young NS. JAMA 2003

Scheinberg P, Nunez O, Young NS. Br J Haematol 2006

CAMPATH IMMUNOSUPPRESSION FOR RELAPSED SAA

Treatment Overall response

Campath (N=25) 14 (56%)

Scheinberg et. al, Blood 2012

For relapsed SAA treatment

with either rabbit ATG or

Campath rescues 2/3 of

patients

Conclusion/Take home points

SAA is a rare disease

SAA is not cancer

Treatments are effective but do not

work in 100% of patients

Conclusion/Take home points

Immunosuppression works in 2/3

of patients and is less toxic than

stem cell transplant but it is not

curative (disease can relapse)

Better immunosuppression

treatments might be coming soon

3/23/2015

7

Conclusion/Take home points

If you relapse after treatment or if

you do not respond to

immunosuppression there are

treatment options

Thank you!

New Directions in Aplastic Anemia:

What's on the Horizon

Bogdan Dumitriu, MD

Hematology Branch

National, Heart, Lung and Blood Institute

National Institutes of Health

Pathophysiology of Aplastic Anemia

Stem cells

Hematopoietic

progenitors

Immune attack

Circulating

blood

cells

Stem cells

Hematopoietic

progenitors

Circulating

blood

cells

HEMATOPOIETIC GROWTH

FACTORS AS THERAPY

FOR SAA

Vadhan-Raj S et al, N Engl 1988; 319:1628: GM-CSF pilot

Ganser A et al, Bood 1990; 76;1287: IL-3 pilots

Kojima S et al, Blood 2002;100:786: G-CSFmonosomy 7

Tichelli A et al, Blood 2011; 117:4434: G-CSF shows no survival

benefit

THROMBOPOIETIN

• Principal endogenous regulator of platelet production.

• Binds to megakaryocyte promotes production of platelets and mature megakaryocytes.

• Stimulatory effects on multilineageprogenitors in vitro and in animal models.

• Clinically maybe beneficial to treat patients with thrombocytopenia (post-chemo, iTP)

3/23/2015

8

Clinical development of THROMBOPOIETIN

• 1994-cloned by 5 independent groups

• 1996-clinical trials with mammalian cell-line produced or

e. coli-produced full length thrombopoietin

– Clearly stimulated platelet production in laboratory animals,

normal volunteers given single doses

– Not particularly effective to speed platelet recovery post-

chemotherapy

• 1999-2000 Serious adverse events reported in platelet

donors

– Normal platelet donors given repeated subcutaneous doses

developed profound, long-lasting thrombocytopenia

– Due to stimulation of cross-reacting autoantibodies against

endogenous TPO (Li et al, BLOOD, 2001)

– Ended clinical development of recombinant thrombopoietin

2nd generation TPO agonists

– Romiplostim (Nplate): IV/SC, chronic

refractory ITP

– Eltrombopag (Promacta): oral, chronic

refractory ITP

ELTROMBOPAG FOR REFRACTORY

SEVERE APLASTIC ANEMIA

Eltrombopag 50 mg daily

Dose escalation every 2 weeks to

150 mg daily

Hematologic responseat 3 months

Non-responders off study

Responders followedmonthly, on drug

NIH Protocol 09-0H0154; ClinicalTrials.gov identifier: NCT00922883

Hematologic Response Criteria

• Platelets: >20K/uL increase, or transfusion-independence

• RBCs: > 1.5 g/dL increase in Hb, or transfusion-independence

• ANC: >100% increase if severe neutropenia, or >500/uL increase

• SAA with plts < 30K/uL

• Refractory to ATG/CSA

PATIENTS CHARACTERISTICS

Age: median, range 44, (18-77)

Male: number, (%) 14, (54%)

Transfusion Dependent Number, (%):

Platelets 26, (100%)

Red blood cells 23, (88%)

Baseline Parameters Median, range

Platelets (K/uL) 9, (5-15)

Neutrophils (K/uL) 0.8, (0.07-2.8)

Hemoglobin (g/dL) 8.0, (6.0-13.8)

Months from last IST Median, range: 12, (6-55)

REFRACTORY SAA ELTROMBOPAG STUDY RESULTS

11 responders (44%)

• 9 platelet responses

• 2 hemoglobin responses

• additional 4 at > 16wks

• 4 neutrophil responses

• additional 3 at > 16wks

26 patientsenrolled

25 evaluablepatients

1 patient ineligible, not treated

14 non-responders

• 10 stable disease

• 2 died of progression

• 2 clonal evolution to MDS

• 1 died

• 1 HSCT

Median follow up 13 months(range 4-28 months)

Censure date 11/1/2011

Pla

tele

ts,

X 1

000/

ul

0

20

40

60

80

100

120

140

1 2 3 4 5 6 7 8 9 10 11

Pt 1

Pt 2

Pt 4

Pt 5

Pt 12

Pt 13

Pt 22

Pt 25

Pt 26

Time in Months

PLATELET RESPONSES

BL 3 6 9 12 15 18 21 24 27 30

3/23/2015

9

Hem

ogl

ob

in,

gr/d

L

Time, months

6

7

8

9

10

11

12

13

14

15

16

17

1 2 3 4 5 6 7 8 9 10 11

Pt 1

Pt 2

Pt 4

Pt 5

Pt 13

Pt 25

HEMOGLOBIN INCREASES AFTER ELTROMBOPAG

Median hemoglobin increase 3.6 g/dL (range 1.5-8.2 g/dL)

BL 3 6 9 12 15 18 21 24 27 30 Ab

solu

te N

eu

tro

ph

il C

ou

nt

X 1

00

0/u

l

Time, months

0

1

2

3

4

5

6

1 2 3 4 5 6 7 8 9 10 11

Pt 1

Pt 2

Pt 4

Pt 5

Pt 12

Pt 13

Pt 20

Pt 24

Pt 25

NEUTROPHIL INCREASES AFTER ELTROMBOPAG

Median ANC increase 590/dL (460-990/uL)

BL 3 6 9 12 15 18 21 24 27 30

BONE MARROW CELLULARITY AT ONE YEAR

Pre-treatment Post-treatment Pre-treatment Post-treatment

Pre-treatment Post-treatment Post-treatmentPre-treatment

RESPONSE SUMMARY OF EXPANDED COHORT

17 responders (40%)

•11 platelet responses

• 4 erythroid responses

• additional 7 at >

16wks

• 8 neutrophil responses

• additional 3 at >

16wks

44 patients

enrolled

43 evaluable

patients

1 patient ineligible,

not treated

26 non-responders

•2 responded >16 weeks

•1 died of progression

•3 deaths from sepsis

•6 clonal evolution

Median follow up 9 months

(range 3-47 months)

1 patient ineligible

not treated

16

Platelets

Neutrophils

Hemoglobin

4

3

3

4

7

12 Weeks-Primary Endpoint Best Response at Follow-up

LINEAGE CHARACTERISTICS OF RESPONSES

2 2 2

1

INSIGHTS INTO SAA PATHOPHYSIOLOGY

FROM ELTROMBOPAG RESPONSIVENESS

stem cell number

probability of failure

correlation with blood counts, age, telomere length

probability of recovery

HSC GROWTH FACTOR (+)

immune attack IST (-)

3/23/2015

10

SUMMARY

• Eltrombopag can promote tri-lineage hematopoiesis in

SAA patients refractory to IST

– 44% clinical response rate

– Transfusion independence

– Well-tolerated

• Eltrombopag stimulation may expand the HSC pool in

humans

• Addition of Eltrombopag early in SAA may increase

response rate, decrease time to response, prevent HSC

depletion, and avoid clonal progression

FDA Approval of Eltrombopag (Promacta) for Refractory Aplastic Anemia

ELTROMBOPAG FOR MODERATE AA

NHLBI 09-H-0154

Eltrombopag, dose escalation to 150 mg QD by mouth

>18 years old; platelet count <30,000/uL

Assessment by blood counts and BM at 3 and 6 months

Horse ATG + CSA and ELTROMBOPAG

for treatment-naïve SAA

NHLBI 12-H-0150

Add eltrombopag to existing horse ATG + CSA platform will

increase overall response and decrease relapses

ANDROGEN THERAPY FOR APLASTIC ANEMIA

May/90Feb/92

16%

48%

38 y/o liver

transplant

26 y/o AA

47 y/o

macrocytosis

21 y/o

macrocytosis

71 y/o

thyroid disease

44 y/o

androgen-

responsive

AA

died age 33 yr

MDS/AML

26 y/o 18 y/o

4 y/o

died age 65 yr “blood

disease” with pallor

wt hTERT

heterozygous K570N

not tested

“SHANK’S DISEASE” IN A MENNONITE FAMILY

23 y/o

dead

26 y/o dairy farmer

progressive pancytopenia

no response to CSA, hormones

HSCT from sister

minimal GVHD, full recovery

TELOMERES AND BONE MARROW FAILURE

3/23/2015

11

TELOMERE STRUCTURE AND BIOLOGY

-Cap chromosome ends

-Tandem TTAGGG repeats

-Bound to array of proteins:

telomerase complex

-Forms higher order chromatin T loop

-Shields 3’ end to prevent recognition

as a DNA “break” by non-homologous

end joining machinery

-TTAGGG loss with proliferation: “end

replication problem”

Autosomal Dominant DKC

Mutations in TERC:

RNA component of the telomerase

complex, the template for telomere

elongation

X-linked DKC

Mutations in DKC1:

encodes dyskerin, a protein

component of telomerase complex

leukoplakia

hyperpigmentation

nail

dystrophy

Courtesy by B. Alter, NCI

TELOMRES AND BONE MARROW FAILURE

DYSKERATOSIS CONGENITA

0

2

4

6

8

10

12

14

16

0 20 40 60 80 100

controls

age, years

telo

me

re le

ng

th, kb

His 412 Tyr

Val 694 MetAla 202 Thr

Cys 772 Tyr

Val 1090 Met

patients

TELOMERE LENGTH IN TERT MUTATION LEUCOCYTESLATE PRESENTATION OF DYSKERATOSIS CONGENITA

37 y/o US Army officer in Afghanistan

tongue ulcer, diagnosed as squamous cell carcinoma

single round of chemotherapy and radiation resulted in unexpected extreme,

persistent pancytopenia. Later, pulmonary metastases

novel Val329Gly mutation in DKC1

Peripheral Blood Telomere Length

0 10 20 30 40 50 60 70 80 90 100 1100.0

2.5

5.0

7.5

10.0

12.5

15.0

Healthy subjects

1952

Age (yr)

Te

lom

ere

le

ng

th (kb

)

Thrombocytopenia, gray hair, very short telomere, TERT mutation

389 C/T (130 Ala/Val)

Telomere disease spectrum

• Moderate to severe aplastic anemia (low platelets only)

• Liver cirrhosis

• Lung fibrosis/IPF

• Skin/nail changes

• Skin/oral cancers

• Leukemia

3/23/2015

12

SEX HORMONES INCREASE TELOMERASE ACTIVITY

IN CULTURED HUMAN LYMPHOCYTES

(n=10)

900

600

Telo

mera

se A

ctivity

(TP

G u

nits)

Methyltrienolone

(synthetic)

300

0

Nandrolone 6β-Hydroxy-

Testosterone

β-Estradiol

0 0.5 5μM 0 5μM 0 5μM 0 1μM

Androgens

Calado RT et al, Blood 2009

Androgens for telomeropathy

Androgens for telomeropathy DANAZOL FOR TELOMEROPATHIES

11-H-0209: “Danazol for Genetic Bone Marrow and Lung Disorders”

Danazol, 800 mg/d x 2 yrs for patients with short telomeres +/- mutations

Phase I/II design, N=31

Primary biologic end point: telomere maintenance/elongation

Secondary clinical end points: toxicity; efficacy (blood counts/PFT and CT)

Protocol opened in August 2011; 27 patients enrolled to date

Modest drug toxicity (minimal LFTs, mild headaches, muscle cramps)

ClinicalTrials.gov identifier: NCT01441037

Conclusion/Take home points

Eltrombopag works as a stem cell

booster

Addition of Eltrombopag to

standard hATG+Csa will likely

improve response rate and

decrease long-term complications

Conclusion/Take home points

10% of aplastic anemia might be

inherited (genetic cause)

Important to diagnose:

- Treatment implications

- Family/genetic counseling

3/23/2015

13

Thank you!

NCT01623167“Eltrombopag with Standard Immunosuppression for Aplastic Anemia”

Indication: SAA

Exclusions: Prior Treatment (ATG, high dose cyclophosphamide, alemtuzumab),

Fanconi Anemia, liver cirrhosis

Inclusions:

• > 2yrs

• Bone marrow cellularity < 30%

• At least two of the following:

ANC < 500/microL

Platelets < 20,000/microL

ARC < 60,000/microL

Purpose: To improve hematologic response by adding eltrombopag (oral growth

factor) to standard immunosuppressive therapy

Type: Inpatient and Outpatient, Non-Randomized

Duration: 10-14 days inpatient for horse-ATG, CSA+ eltrombopag for 6 months, with 3

month, 6 month and annual follow-up visits

Severe Aplastic Anemia (SAA)Treatment-Naive

NCT00922883 “ A Pilot Study of a Thrombopoietin-receptor Agonist (TPO-R agonist),

Eltrombopag, in Aplastic Anemia Patients with Immunosuppressive-therapy Refractory

Thrombocytopenia”

Indication: SAA with refractory thrombocytopenia following at least one treatment course of

horse or rabbit ATG/cyclosporine.

Exclusions: Treatment with horse or rabbit ATG or Campath within 6 months of study entry,

Fanconi Anemia, liver cirrhosis, PNH clone size greater than 50%.

Inclusions:

• Diagnosis of aplastic anemia, with refractory thrombocytopenia following at least one

treatment course of horse or rabbit ATG/cyclosporine.

• Platelet count ≤ 30,000/mL

• Age ≥ 18 years old

Purpose: To improve platelet counts with eltrombopag (oral growth factor) in patients with

thrombocytopenia after immunosuppressive therapy

Type: Outpatient, Non-Randomized

Duration: 3 months of therapy. Monthly visits to NIH.

Severe Aplastic Anemia (SAA)Refractory

NCT01328587 “Eltrombopag for Moderate Aplastic Anemia”

Indication: MAA with thrombocytopenia or anemia.

Exclusions: Fanconi Anemia, liver cirrhosis, PNH clone size greater than 50%.

Inclusions: Current diagnosis of moderate aplastic anemia and

• Age ≥ 18 years old

• One or more of platelet count less than or equal to 30,000/microL, Hemoglobin less

than or equal to 8.5g/dl OR platelet and/or red cell transfusion dependent

Purpose: To improve platelet counts with eltrombopag (oral growth factor).

Type: Outpatient, Non-Randomized.

Duration: 4 months of therapy. Monthly visits to NIH.

Moderate Aplastic Anemia (MAA)

NCT00345345 “Alemtuzumab to Treat T-Large Granular Lymphocytosis”

Indication: T-LGL, Neutropenia, Anemia

Inclusions:

1. T-LGL clone detectable by peripheral blood flow cytometry and TCR gene

rearrangements

2. At least one cytopenia:

• ANC < 500/uL

• Platelets < 20 k/uL or < 50 k/uL with bleeding

• Hemoglobin < 9 g/dL or anemia requiring >2 units of RBCs/month transfusions

Purpose: To characterize response (improvement of blood counts) to immunosuppression

(Campath) in patients with T-LGL

Type: Inpatient and Outpatient, Non-Randomized

Duration: 11 days inpatient, with 3 month, 6 month and annual follow-up visits

T-cell Large Granular Lymphocytosis (T-LGL)

• A Pilot Study of a Thrombopoietin-Receptor Agonist, Eltrombopag, in Patients With Low to Int-2 Risk MDS

• NCT00961064

• Purpose of this study is to see if eltrombopag can increase platelet counts in patients with MDS

• Inclusion criteria:– Patients 18 years of age and older who have platelet count of less than 30

that has not responded to conventional treatment

• Outpatient study: – Treatment with eltrombopag once per day for 16 weeks

– Evaluations at NIH every 4 weeks

– If response, then follow up every 3 months

– If no response then follow up at 1 and 6 months off drug

Myelodysplastic syndrome (MDS)

3/23/2015

14

Clofarabine followed by high-dose lenalidomide for high risk MDS, CMMoland AML

NCT01629082

• PhaseI

• Single course of IV clofarabine (5mg/m2/d x5) for cytoreduction

• Oral lenalidomide consolidation with dose escalation from 25 mg daily for 21/28 days for 1 cycle in the first cohort up to 50 mg daily for 28/28 days for 2 cycles in the fourth cohort.

• Oral lenalidomide maintenance, starting at a dose of 10 mg daily in 28 day cycles, with dose adjustments, for up to 12 cycles.

• Inclusion criteria:

– MDS: Int-2 IPSS and higher

– CMMoL

– AML

– ECOG 0 to 2

– Unresponsive to, or unable to receive, DNA hypomethylating agents or standard intensive induction chemotherapy (i.e. “7+3” cytarabine/anthracycline chemotherapy)

• Outpatient study:

– Needs to stay locally for monthly revlimid prescription refills.

Myelodysplastic syndrome (MDS)