HO Questions Flashcards

1
Q

What is the single strongest prognostic factor in acute lymphoblastic leukemia (ALL)?
a. Age
b. Initial WBC count
c. Sex
d. Early treatment response as assessed by minimal residual disease (MRD) at end of induction therapy
e. Presence of CNS leukemia (CNS3)

A

Answer D is correct. Multivariate analyses consistently show that end induction MRD is the strongest prognostic factor in ALL. Age (answer A) and initial WBC count (answer B) remain prognostic but are not as powerful as MRD in multivariate analyses. Sex (answer C) remains prognostic of outcome in very large trials, with boys having inferior outcome to girls; however, sex is of only limited prognostic significance, with relative risks of 1.1 to 1.2 (which cannot be detected unless a very large number of patients is analyzed). CNS involvement (answer E) is prognostic of outcome in some but not all contemporary trials. It is more important as an indicator of a need for more CNS-directed therapy than as a prognostic factor.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

Novel immunotherapeutics currently are being used in acute lymphoblastic leukemia (ALL) therapy, including bispecific T-cell engaging therapies. Cytokine release syndrome is a known complication of this therapy. What cytokine is associated with the acute onset of the inflammatory response seen in this acute complication?
a. IL-7 b. C3 c. IL-6 d. IL-4 e. IL-3

A

Answer C is correct. Abnormal macrophage activation can occur in the context of blinatumomab therapy, particularly in the setting of high disease burden. Cytokine release syndrome (CRS) is best managed with supportive care. Laboratory values consistent with hemophagocytic lymphohistiocytosis include elevated ferritin, cytopenias, and hypofibrinogenemia. Elevated levels of IL-6 are associated with CRS and the use of tocilizumab, an IL-6 receptor antibody, can ameliorate the symptoms. IL-7 (answer A) is a signaling molecule involved in B-cell differentiation. C3 (answer B) is not generally assessed in CRS. IL-4 (answer D) induces differentiation of naive helper T cells to Th2 cells. IL-3 (answer E) promotes development of myeloid progenitor cells through binding of the IL-3 receptor.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

A 17-year-old boy presents with a 1-week history of cough and increasing shortness of breath. His pediatrician notes decreased breath sounds bilaterally and obtains a chest x-ray, which shows an anterior mediastinal fullness. He is transported to your hospital and has a CBC showing a white blood cell count of 180,000/dL with less than 50% circulating blasts. His coagulation parameters are also abnormal, with an international normalized ratio (INR) of 1.8 and a partial thromboplastin time (PTT) of 40 seconds. Immunophenotyping demonstrates an early T-cell precursor subtype. Which of the following characteristics portends the worse prognosis for this patient?
a. High presenting white blood cell count
b. Age at presentation
c. Abnormal coagulation tests
d. Persistent disease at the end of consolidation therapy
e. Early thymic precursor (ETP) immunophenotype

A

The correct answer is D. There is no prognostic significance to age or presenting white blood cell count for patients with T-cell acute lymphoblastic leukemia (ALL). It is common to see anterior mediastinal masses in T-cell ALL, and coagulopathies are common and can sometimes cause delays in diagnostic lumbar punctures. When first reported, the early thymic precursor (ETP) phenotype was considered a poor prognostic factor in retrospective studies, but more recently presented data do not support that ETP ALL represents a subtype of T ALL with a worse prognosis. However, as with other ALL subtypes, response to therapy is one of the most important predictors of future relapse. For T-cell ALL, end induction (also known as time point 1 in some European studies) minimal residual disease is not as prognostic as end consolidation (also known as time point 2 in some European studies).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

A newly diagnosed patient with acute lymphoblastic leukemia (ALL) has a twin. Which of the following circumstances are associated with the highest risk for ALL development in that twin?
a. A 3-year-old boy with ALL and ETV6-RUNX1 (TEL-AML1) fusion who has an identical twin brother b. A 6-month-old boy with ALL and KMT2A::AFF1 (KMT2A::AF4) fusion who has a twin sister
c. A 6-year-old boy with ALL and MLL-AFF1 (MLL-AF4) fusion who has an identical twin brotherA 6-year-old boy with ALL and KMT2A::AFF1 (KMT2A::AF4) fusion who has an identical twin brother
d. A 6-month-old boy with ALL and KMT2A::AFF1 (MLL::AF4) fusion who has an identical twin brother
e. A 2-year-old boy with ALL and TCF3::PBX1 (E2A::PBX1) fusion who has an identical twin brother

A

Answer D is correct; the concordance rate for leukemia is highest for identical twins diagnosed in the first year of life. The concordance rate is about 50% in the first year of life and drops to a very low percentage by age 5 years. Concordance occurs because of in utero twin-to-twin transfer of either overt leukemia cells or cells with an initiating leukemia event (typically a translocation), with new secondary events acquired independently in each twin. Because 75% to 80% of infants with ALL have KMT2A translocations, most but not all cases of concordant ALL in twins are associated with KMT2A translocations. ETV6::RUNX1 and KMT2A translocations typically occur in utero in patients diagnosed with leukemia in the first few years of life; the identical twins of the patients in answers A and C are at some risk at 3 (A) and 6 (C) years, but probably only 5% to 10% for A and a very low risk for C. The twin of the patient in answer B is fraternal and at much lower risk than the identical twin in answer D. TCF3::PBX1 is much less likely to occur in utero than ETV6::RUNX1 and KMT2A translocations, so the twin of the patient in answer E is at low but not zero risk.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

A mother brings her 3-year-old son to the emergency department for excessive bruising. She gave him a bath before bedtime on Sunday night, and he was fine. On Monday morning, when she dressed him, she noticed tiny red spots on his skin. During the day, he developed extensive bruising without known trauma. He is otherwise well, has no fever, and has been playing happily. He is described as being very active and always climbing on things. He has a prior history of a humerus fracture at 18 months and a femur fracture at 30 months.
What is the most likely explanation for this history?
a. Acute lymphoblastic leukemia (ALL)
b. Acute myeloid leukemia (AML)
c. Nonaccidental trauma
d. Idiopathic thrombocytopenic purpura (ITP) e. Viral infection

A

Answer D is correct. In cases of ITP, the parents often can describe exactly when the onset of petechiae and bruising occurred, and children typically are otherwise well. ALL and AML (answers A and B) are possible, but the acute onset of petechiae and bruising is evidence against these diagnoses. In addition, most children with newly diagnosed acute leukemia have other complaints such as bone pain, lethargy, irritability, and fever. Although the prior history of fractures raises concern about nonaccidental trauma (answer C), the factors above argue against this diagnosis, and a CBC should reveal isolated thrombocytopenia. Viral infections (answer E) can be associated with cytopenias, but one would not expect the child to be otherwise well.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

The National Cancer Institute (NCI)/Rome risk factors are used to group patients with acute lymphoblastic leukemia (ALL) into standard- and high-risk groups. Which of the following patients has standard-risk ALL?
a. A 9-year-old boy with WBC count 45,000/μL and B-lineage ALL
b. An 11-and-a-half-month-old girl with WBC count 5,000/μL and B-lineage ALL c. An 11-year-old girl with WBC count 5,000/μL and B-lineage ALL
d. A 13-year-old boy with WBC count 5,000/μL and T-cell ALL
e. A 3-year-old boy with WBC count 55,000/μL and B-lineage ALL

A

Answer A is correct. Standard-risk patients are those with age 1.01 to 9.99 years and initial WBC count less than 50,000/μL. Patient B is an infant younger than 1 year. Patient C has high-risk ALL due to age greater than 10 years. The NCI criteria apply only to B-lineage ALL, and patient D has T-ALL. Patient E has high-risk ALL based on a WBC count greater than 50,000/μL.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

A 13-year-old girl with relapsed acute lymphoblastic leukemia (ALL) is undergoing reinduction chemotherapy. She develops high fevers with neutropenia approximately 3 weeks into her course, and a CT scan of her chest demonstrates four isolated pulmonary nodules that are about 1 cm in dimension. Her galactomannan is positive. What is the most appropriate antimicrobial coverage for her?
a. Caspofungin
b. Voriconazole
c. Fluconazole
d. Nystatin

A

Answer B is correct. The empiric therapy of pulmonary nodules during febrile neutropenic episodes has been revolutionized by the availability of voriconazole, a triazole antifungal medication. Caspofungin is second-line for aspergillosis, fluconazole has no activity against aspergillus, and Zosyn or another antibiotic would have already been in use in this patient.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

Which of the following patients is expected to have the best prognosis?
a. A 3-year-old boy with an initial WBC count of 2,000/mcL, ETV6-RUNX1 (TEL-AML1) fusion, and minimal residual disease (MRD) 0.14% at end induction
b. A 3-year-old girl with an initial WBC count of 2,000/μL, TCF3::PBX1 (E2A::PBX1) fusion, and MRD 0.04% at end induction
c. A 3-year-old girl with an initial WBC count of 9,000/μL, ETV6::RUNX1 (TEL::AML1) fusion, and MRD 0.07% at end induction
d. A 3-year-old girl with an initial WBC count of 2,000/μL, hyperdiploidy with trisomies of chromosomes 4 and 10, and MRD 0.04% at end induction
e. A 3-year-old girl with an initial WBC count of 9,000/μL, hyperdiploidy with trisomies of chromosomes 4 and 10, and MRD less than 0.01% at end induction

A

Answer E is correct. Age, sex, initial WBC count, blast cell genetics, and day 29 MRD response all are prognostic of outcome. Patients aged 2 to 6 years with a WBC count less than 10,000 have the best prognosis. All of the patients in this example fall into this category. Initial WBC is a continuous variable, but it is hard to parse out the difference between 2,000/μL and 9,000/μL. The most favorable genetic features are ETV6::RUNX1 (TEL::AML1) fusion (present in answers A and C) and hyperdiploidy with favorable chromosomes trisomies (with 4 and 10 being most important; present in answers D and E). However, MRD response is the strongest prognostic factor, with patients with MRD less than 0.01% clearly doing best (present only in answer E).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Which of the following characteristics excludes a diagnosis of B-cell precursor acute lymphoblastic leukemia (ALL)?
a. Presence of T-cell receptor gene rearrangements
b. Expression of cytoplasmic CD3
c. Expression of surface CD13/33
d. Mediastinal mass
e. WBC count greater than 500,000/μL

A

Answer B is correct. Cytoplasmic CD3 expression is a diagnostic criterion for T-cell acute lymphoblastic leukemia (T-ALL) and cannot be present in B-cell precursor ALL. T-cell receptor gene rearrangements (answer A) occur in B- precursor ALL and are often used to monitor minimal residual disease. CD33 expression (answer C) is common in B-cell precursor ALL. Both a mediastinal mass (answer D) and hyperleukocytosis (answer E) are much more common in T-ALL than in B-cell precursor ALL, but neither precludes a diagnosis of B-cell precursor ALL.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Which of the following factors is most prognostic for the highest-risk subgroup of infants younger than 1 year with acute lymphoblastic leukemia (ALL)?
a. CNS involvement
b. Initial WBC count 50,000/μL
c. NUTM1 translocations
d. Age younger than 3 months
e. KMT2A translocations and age younger than 3 months

A

Answer E is correct. The strongest prognostic factors in infants with ALL are age (younger than 3 months is worse than 3 months to younger than 6 months, which is worse than 6 to 12 months) and the presence of a KMT2A translocation. Infants with both age younger than 3 months and KMT2A translocations have an extremely poor outcome. CNS involvement (answer A) is much more common in infants with ALL than in older children, does not have prognostic significance. The average WBC count is much higher in infants than in older children with ALL, and most have a WBC count of 50,000/μL or greater (answer B). This (answer B) is of prognostic importance but is nowhere near as strong as age less than 3 months and MLL translocations. Some studies (Interfant-99) have found that WBC 300,000/μL or higher is a particularly poor prognostic marker. More recently, infants with NUTM1 alterations are found to have more favorable prognoses.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

A 16-year-old Hispanic boy presents with fever, fatigue, and swollen glands. A CBC demonstrates a WBC count of
89,000/µL and cytogenetics revealed a t(Y;14)(p11;q32). Which of the following fusion genes is most likely to be
present?
a. MLL-AF9
b. BCR-ABL1
c. IGH-CRLF2
d. TCF3-PBX1
e. EWS-FLI1

A

Answer C is correct. The presence of a chromosome translocation in most or all cells generally is indicative of a
malignancy, although balanced translocations (Robertsonian translocations) can be seen in individuals without
malignancies. The IGH/CRLF2 rearrangement indicative of Ph-like acute lymphoblastic leukemia (ALL) is more
common in Hispanic adolescents and is associated with high expression of CRLF2. The locations of the other
fusion genes are not on chromosome 14 or chromosome Y. EWS-::FLI1 (answer E) is a fusion gene found in Ewing
sarcoma.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

Which of the following chromosome translocations is most likely to be seen in pediatric T-cell acute lymphoblastic
leukemia (T-ALL)?
a. t(9;22)(q34;q11)
b. t(8;22)(q24;q11)
c. t(1;19)(q23;p13)
d. t(9;11)(q34; q23)
e. t(11;14)(p13;q11.2)

A

Explanation
Answer E is correct. The t(11;14)(p13;q11.2) fuses the T-cell receptor alpha/delta (TCRA/D) locus with LMO2,
which occurs in about 7% of pediatric T-ALL cases and does not appear to have prognostic significance. The
t(9;22)(q34;q11) or Philadelphia chromosome (answer A) occurs in about 4% of pediatric B-cell precursor ALL.
The Philadelphia chromosome is also seen rarely in T-ALL, but with a frequency less than 1%. The
t(8;22)(q24;q11) fuses c-MYC to the immunoglobulin lambda gene on chromosome 22 (answer B) and is a rare
recurrent translocation in Burkitt leukemia and lymphoma, not T-ALL. The t(1;19)(q23;p13) (answer C) creates
TCF3::PBX1 (E2A::PBX1) fusion and is seen in about 5% of B-cell precursor ALL cases. The t(9;11)(q34;q23)
(answer D) creates KMT2A::AF9 fusion and is seen commonly in acute myeloid leukemia and less commonly in Blineage ALL (especially infants).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

A 15-year-old male with B-lineage ALL receives induction therapy. On day 24, he begins complaining of
abdominal pain and nausea. Serum lipase is 940 U/L, TB 3.0, DB 2.0, and AST 320. Abdominal ultrasound
demonstrated a swollen pancreas with some cystic structures. What induction chemotherapy likely caused this
constellation of findings?
a. Vincristine
b. Cytarabine
c. Asparaginase
d. Daunomycin
e. IT Methotrexate

A

he answer is C, asparaginase. Asparaginase is a key agent used to treat ALL. It depletes asparagine, a nonessential amino acid, in both lymphoblasts and healthy cells. While the exact mechanism of how asparaginase
causes pancreatitis is not fully known, there is a higher incidence of pancreatitis secondary to asparaginase in
older patients.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

A 12-year-old boy has T-cell acute lymphoblastic leukemia (ALL) with an initial WBC count of 500,000/µL and
CNS3 status with CSF WBC 200/µL (100% blasts) and RBC 10/µL. The treatment regimen specifies that CNS
radiation should be given.
What would the proper radiation regimen consist of?
a. 2,400 cGy to whole brain, excluding the globes of the eyes, and 1,200 cGy to spine
b. 1,800 cGy to whole brain, excluding the globes of the eyes, and 1,200 cGy to spine
c. 1,800 cGy to whole brain, excluding the globes of the eyes
d. 1,800 cGy to whole brain, including the entire globes of the eyes
e. 1,800 cGy to whole brain, including the posterior halves of the globes of the eyes

A

Answer E is correct. Radiation therapy for ALL no longer routinely includes spinal radiation, which eliminates
answers A and B. The standard dosage for patients with clear CNS involvement (as present in this case) generally
is considered to be 1,800 cGy, with the target volume including the entire brain and meninges, including the
frontal lobe and posterior halves of the globes of eyes, with optic disc and nerve, extending superior to vertex
and posterior to occiput. Caudal border will be below the skull base at C2 vertebral level. Some groups (St. Jude,
UKALL, and DCOG) now believe that even patients with CNS3 involvement do not need CNS irradiation, but this is
not the majority opinion at this time.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Which of the following patients should not be treated with therapy used commonly for pediatric acute
lymphoblastic leukemia (ALL)?
a. A 10-year-old boy with a large mediastinal mass, pleural and pericardial effusions, normal peripheral
blood cell counts, and 3% T-lymphoblasts in the bone marrow
b. A 4-year-old girl with L2 morphology ALL with lymphoblasts expressing cytoplasmic mu heavy chains
c. A 3-year-old boy with ALL that expresses CD10, CD19, CD13, and CD33
d. A 20-year-old woman with B-precursor ALL and a white blood cell count of 40,000/µL
e. A 4-year-old boy with WBC count 45,000/µL, hepatosplenomegaly, and 50% lymphoblasts with deeply
basophilic cytoplasm and cytoplasmic vacuoles, surface kappa light chains, and a t(2; 8)(p12;q24)

A

Answer E is correct. Burkitt leukemia is defined by L3 morphology, with deeply basophilic cytoplasm and
cytoplasmic vacuoles and the presence of translocations that join the c-Myc locus at 8q24 to an immunoglobulin
heavy or light chain gene. The immunoglobulin kappa gene is located at 2p12. These patients require different
therapy than other patients with ALL and typically are treated in the same way as patients with advanced stage
Burkitt lymphoma using therapy that is intensive but short in duration and not including ALL maintenance
chemotherapy. The patient described in answer A has stage III T-cell lymphoma, which is typically treated the
same as is T-cell ALL. The patient described in answer B has pre-B cell ALL, and the patient described in answer C
has ALL with expression of myeloid markers. The latter is not unusual and not associated with an adverse
prognosis, so he should be treated like any other patient with ALL. The young woman described in answer D will
fare much better if treated on a pediatric rather than adult ALL protocol.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

Your patient with acute myeloid leukemia (AML) is receiving an intensive course of chemotherapy with high-dose cytarabine. She has been neutropenic for 4 days, has developed severe mucositis, and is now hypotensive.
Which of the following is most likely to be isolated from this patient’s blood culture?
a. Pneumocystis jiroveci
b. Staphylococcus epidermidis c. Aspergillus fumigatus
d. Herpes simplex virus (HSV) e. Streptococcus viridans

A

Children with AML are at high risk of developing bacteremia and specifically S. viridans, also known as alpha hemolytic strep. The incidence increases significantly with mucositis or high-dose cytarabine in patients with AML, and they should be covered with empiric antibiotics used in each institution’s neutropenic fever guidelines. S. epidermidis is a common cause of bacteremia but is rarely associated with hypotension. Aspergillus can be seen in this population but typically in patients with longer periods of neutropenia, and it typically does not cause hypotension as a presenting symptom. Although HSV is associated with mucositis, hypotension is not a characteristic finding. Pneumocystis is not a common complication of AML therapy and is not specifically linked with this scenario. Other organisms with similar presentation not listed above include Staphylococcus aureus, and the gram-negative organisms and empiric antibiotic choices should also be directed against these organisms.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

A 5-year-old boy presents with acute myeloid leukemia (AML) and a WBC count of 120,000/mm3. Cytomolecular genetics reveals a NUP98 fusion and all other testing is negative. She is treated with 10 days of daunorubicin, AraC, and etoposide for induction therapy. On day 30, a bone marrow aspiration shows 30% leukemic blasts. She enters remission after treatment with idarubicin, fludarabine, and high-dose AraC. She has no HLA-matched siblings, but an unrelated donor search reveals a large number of potential matches.
Which course of treatment is most likely to result in the best outcome?
a. Give one additional course of induction therapy followed by three more courses of intensification chemotherapy.
b. Perform an autologous hematopoietic stem cell transplant (HSCT) now.
c. Give one more course of intensification chemotherapy and then perform a matched unrelated donor
HSCT.
d. Give one more course of intensification chemotherapy and then 1 year of maintenance chemotherapy.

A

Patients with intermediate-risk AML who have residual leukemia after the first induction course have been shown to have a high risk of relapse with chemotherapy alone. Allogeneic HSCT is likely the optimal therapy in this setting. Continuing chemotherapy alone will be very unlikely to result in cure, and maintenance chemotherapy is not a standard component of most AML treatment protocols. Similarly, autologous HSCT is also unlikely to provide a curative approach to therapy, because of the potential for contaminating leukemia cells in the graft and the lack of a graft versus leukemia effect. Because it will take time to arrange an unrelated allogeneic donor, giving another course of chemotherapy to maintain remission followed by HSCT would be the best of the available treatments.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

A family requests consultation with you after their 2-year-old boy with trisomy 21 recently is diagnosed with acute myeloid leukemia (AML). They are struggling with a decision whether to have their child treated, and, if they do agree to treatment, what it should include. The initial presentation included findings that the leukemia was positive for CD41 and CD61, fluorescence in situ hybridization was only positive for trisomy 21, the initial white count was 75,000/mm3, the liver and spleen were moderately enlarged, and the child is otherwise well and stable.
,br/>Based on current literature, what do you recommend that the regimen chosen should include?
a. Induction chemotherapy with anthracyclines and cytarabine, at least one course of intensive cytarabine,
and intermittent dosing of intrathecal cytarabine over the course of treatment
b. No therapy, because this will spontaneously resolve c. Low-dose cytarabine alone
d. Induction chemotherapy with anthracyclines and cytarabine, concluding with a bone marrow transplant if a sibling donor is available
e. Because of the poor prognosis in these children with this type of leukemia, only palliative treatment

A

This patient has the typical acute megakaryoblastic leukemia seen in children with Down syndrome. The patient’s age is younger than 4 years. Although the outcome for AML in children with Down syndrome who are younger than 4 years is favorable, it does require therapy, and therefore answers B and E are incorrect. Answer B, observation, is indicated in low- or moderate-risk transient myeloproliferative disorder (TMD), which is restricted to the first 90 days of life, and answer C would be appropriate in high-risk TMD. Studies have shown that the best outcome for a child with this presentation is seen with answer A. Remissions in children with Down syndrome treated with low-dose cytarabine are not sustained. Children with Down syndrome who received a matched sibling transplant actually had a worse outcome than those who received high-dose cytarabine during intensification.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

A 7-year-old girl is found to have a white count of 55,000/mm3 with 20% neutrophils, 10% lymphocytes, and 70% blasts. Bone marrow aspirate and biopsy shows 75% blasts. The blasts on flow cytometry show CD33, CD13, and CD34 to be positive; CD7 to be minimally positive; and TdT, CD3 (surface and cytoplasmic), and CD10 to be negative.
Which of the additional testing options below is most important to further define risk-based therapy in this patient’s leukemia?
a. Cytogenetics/fluorescence in situ hybridization (FISH) for BCR-ABL1, iAMP21, and ploidy
b. Cytogenetics/FISH for inv(16) and t(8;21), and FLT3 mutation testing
c. WBC greater than 50,000/mm3, cytogenetics for ploidy, and immunohistochemistry for myeloperoxidase d. Cytogenetics/FISH for t(11;19) and +21, and sequencing for RAS mutation status
e. Cytogenetics/FISH for t(1;22) and trisomy 8, and sequencing for GATA1 mutation status

A

This patient’s bone marrow aspirate, biopsy, and flow cytometry are diagnostic of acute myeloid leukemia (AML). Therefore, the important prognostic factors on which initial risk stratification is based include favorable cytogenetics and molecular markers (inv(16), t(8;21), NPM1, CEBPA), unfavorable cytogenetics and molecular markers (monosomy 7, monosomy 5 or deletion 5q, FLT3-ITD high allelic ratio), and induction response. The other choices are prognostic for patients with acute lymphoblastic leukemia (answer A) or do not factor into prognostic classifications for AML (answers C through E).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

A 12-year-old girl presents with a WBC count of 750,000/mm3 with 1% blasts and other immature myeloid cells at different stages of differentiation. Her only significant sign or symptom is abdominal pain and a “swollen belly.” The platelet count is 220,000/mm3, and the hemoglobin is 11 g/dL. The uric acid is 3.
What is the most likely diagnosis and optimal treatment for this patient?
a. Chronic myeloid leukemia (CML) in accelerated phase; treat with imatinib and hydroxyurea. b. CML in chronic phase; treat with imatinib.
c. Myelodysplastic syndrome transforming into acute myeloid leukemia (AML); treat with induction chemotherapy followed by family donor ablative bone marrow transplantation once remission is achieved.
d. CML in accelerated phase; treat with imatinib followed by matched family donor bone marrow transplantation.
e. AML; treat with induction chemotherapy.

A

The high WBC with a “normal” maturation of the myeloid lineage in the peripheral blood (including only a small percentage of myeloblasts) along with minimal symptoms, in this case due to splenomegaly, is most characteristic of CML in chronic phase. The normal platelet count and hemoglobin are also consistent with this. Accelerated phase CML is characterized by at least 10% but less than 30% leukemic blasts in the peripheral blood or bone marrow. In addition, thrombocytopenia with a platelet count of less than 100,000/mm3 usually accompanies accelerated phase CML. AML is usually associated with increased blasts, anemia, and thrombocytopenia without the “normal” maturation of the myeloid lineage observed on the peripheral smear. Imatinib (or a “second-generation” tyrosine kinase inhibitor, such as dasatinib or nilotinib) is almost always adequate initial therapy, which will result in an effective cytoreduction in chronic phase CML and, in a majority of cases, leads to a cytogenetic remission, although fewer molecular remissions are obtained. However, allogeneic hematopoietic stem cell transplant (HSCT) remains the only known curative therapy for CML at this time. Until more information about drugs such as imatinib is obtained, in terms of long-term follow-up, the role of allogeneic HSCT is controversial.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

A 15-year-old boy presents for a follow-up visit for localized Ewing sarcoma of the pelvis. He was diagnosed 18 months ago and completed therapy 6 months ago. Therapy included 14 cycles of standard chemotherapy and radiation for local control. He was last seen 3 months ago and had negative scans and normal blood work. He presents today with recent onset of fatigue, a white count of 90,000/mm3, hemoglobin of 6.9 g/dL, platelets of 40,000/mm3, and hepatosplenomegaly. You suspect that he has therapy-related acute myeloid leukemia (AML).
Of the following, which chemotherapy drug is the most likely culprit, and which cytogenetic abnormality is most likely to be present in the leukemia?
a. Doxorubicin; monosomy 7 b. Etoposide; t(9;11)
c. Ifosfamide; t(9;11)
d. Etoposide; monosomy 7 e. Ifosfamide; monosomy 7

A

Therapy-related myeloid neoplasms (therapy-related myelodysplastic syndrome [t-MDS] or t-AML) are a dreaded, and fortunately rare, complication of treatment with chemotherapy. The two major classes of chemotherapy with the highest risk of t-MDS/AML are alkylating agents (eg, cyclophosphamide, ifosfamide) and topoisomerase inhibitors (eg, etoposide, doxorubicin). The t-MDS/AML arising from these two classes of agents typically differs in two important ways. First, t-MDS/AML arising after exposure to alkylators typically has a long latency (3 to 5 years) with a long preleukemic MDS phase, whereas that arising after exposure to topoisomerase II inhibitors typically has a short latency (6 to 18 months) with a more explosive presentation. Second, t-MDS/AML arising after alkylator exposure often carries high-risk cytogenetic lesions such as monosomy 7 or 5q-, whereas cases arising after topoisomerase II inhibitor exposure typically are most likely to harbor an MLL rearrangement, most commonly t(9;11). Given the short latency and rapid progression in this case, answer B contains the most likely etiology and cytogenetic results.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

A 15-year-old girl has just been diagnosed with chronic myeloid leukemia, and you have initiated therapy with the tyrosine kinase inhibitor (TKI) imatinib at 400 mg daily.
What can you anticipate in this case?
a. Complete hematologic and cytogenetic remission within 1 month and resolution of splenomegaly within 6 months; you plan to continue imatinib for a total of 9 months.
b. Complete hematologic remission within 3 months, complete cytogenetic remission (CCyR) within 12 months, and major molecular response (MMR) within 18 months; monitor by peripheral blood RT-qPCR for BCR/ABL every 3 to 6 months while continuing imatinib indefinitely if MMR persists.
c. Complete hematologic remission within 2 weeks and cytogenetic remission within 4 weeks; you will monitor RT-qPCR for BCR/ABL monthly during the 1-year maintenance therapy with imatinib.
d. Complete hematologic remission within 6 months and CCyR within 12 months; after CCyR is achieved, you will discontinue RT-qPCR monitoring.

A

Initial response to TKI therapy is typically gradual and proceeds in the following order: resolution of splenomegaly and normalization of blood counts, cytogenetic remission of the marrow, and diminution of peripheral blood BCR/ABL by RT-qPCR to low or undetectable levels. The expected rate of this resolution is correct in answers B and D. Monitoring for recurrence is recommended by use of peripheral blood RT-qPCR, which is most sensitive. Thus, the correct answer is B. For now, the duration of imatinib or other TKI administration is thought to be indefinite, although ongoing adult long-term follow-up studies that include discontinuation of TKIs will shed more light on this.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

A 3-year-old girl is referred to you for evaluation of thrombocytopenia. She has trisomy 21 and was noted to have transient myeloproliferative disorder as an infant, which resolved spontaneously without the need for chemotherapy. A bone marrow is performed, which demonstrates increased blasts and fibrosis. The immunophenotyping marker that is most likely to be positive is:
a. CD4 b. CD19 c. CD22 d. CD41 e. TdT

A

This patient with trisomy 21 and history of transient myeloproliferative disorder is at significant risk for developing acute myeloid leukemia, specifically acute megakaryoblastic leukemia (AMKL). Twenty percent of patients with Down syndrome and TMD will develop AML, which typically presents in the first 3 years of life with isolated thrombocytopenia or pancytopenia, and bone marrow fibrosis is often present. AMKL associated with Down syndrome has a superior prognosis compared to non-Down AMKL and is treated with less intensive chemotherapy than traditional AML. Answer A is incorrect because CD4 is associated with T-ALL. Answers B and C (CD19 and CD22) are B-cell markers, and answer D (TdT) is an immature lymphoid marker. The correct answer (D) is CD41, which is a megakaryocytic marker characteristic of AMKL, as are CD42 and CD61. Although patients with Down syndrome also have an increased lifetime risk of developing ALL, this question seeks to reinforce the association between TMD and AMKL in Down syndrome.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

You have a new 15-year-old male patient with a white count of 28,000/mm3 and 11% myeloblasts, hemoglobin of 7.2 g/dL, and platelet count of 30,000/mm3. A bone marrow aspirate reveals 18% blasts that have Auer rods and are surface marker positive for CD33. You receive a call from the cytogenetics lab that the bone marrow karyotype is positive for t(8;21) in 17 out of 20 metaphases. Your staff asks whether this represents a diagnosis of acute leukemia in the current classification scheme for this type of hematologic malignancy.
a. No, because for a diagnosis of acute leukemia you must have 30% or more blasts in the marrow. b. No, because for a diagnosis of acute leukemia you must have 20% or more blasts in the marrow. c. No, because the cytogenetics do not include +21, monosomy 7, or trisomy 8.
d. Yes, because Auer rods are present.
e. Yes, because the cytogenetics includes a characteristic AML-defining mutation.

A

The patient has a myeloid neoplasm by virtue of the presenting histochemical findings and cell surface markers. The current classification (ie, World Health Organization [WHO]) uses a minimum of 20% blasts in the marrow for a designation of acute myeloid leukemia (AML) versus myelodysplastic syndrome if there are fewer than 20% blasts. In the old French-American-British classification scheme, this cutoff had been 30% or more for a diagnosis of AML. A crucial feature of the WHO classification is that a patient need not have 20% blasts in the marrow for a diagnosis of AML if they have one of the classic AML cytogenetic findings by conventional karyotype or by fluorescence in situ hybridization (ie, t(8;21), t(15;17), inv(16), 11q23 translocation (KMT2A rearrangements), NPM1 mutations, MECOM, NUP98 rearrangements).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

You are seeing a 19-year-old man who was diagnosed with chronic myeloid leukemia 2 years ago. He has been taking imatinib 400 mg daily since diagnosis. He has no siblings, but there are multiple potential matched unrelated donors in the registry. He has been fully compliant with his imatinib, and side effects have been minimal. Bone marrow and peripheral blood 6 months after the patient began imatinib were negative for BCR- ABL 1 by fluorescence in situ hybridization (FISH), and FISH in peripheral blood has remained negative up until the 21-month check. Peripheral blood qRT-PCR for BCR-ABL 1 was 0.03% (international scale) 1 year after the patient began imatinib and has remained less than 0.1% up until the 21-month check. On 24-month testing, however, peripheral blood FISH is positive at 6% and qRT-PCR is positive at 11.3%. Blood counts remain normal.
What is your next step?
a. Immediately finalize a matched unrelated donor and proceed to transplant.
b. Increase the imatinib dosage to 800 mg daily.
c. Add interferon to imatinib.
d. Send peripheral blood for ABL resistance mutation testing and switch from imatinib to dasatinib.

A

This patient has developed secondary resistance to imatinib after achieving a complete cytogenetic and major molecular response. Although increasing the imatinib dosage has been effective for some patients in this situation, the second-generation tyrosine kinase inhibitors (TKIs) dasatinib and nilotinib are more likely to reestablish an optimal response. ABL sequencing to detect resistance mutations can help guide alternative TKI choice. For example, patients with T315I mutations should receive ponatinib. Although stem cell transplant with a suitable donor may be indicated if alternative TKI therapy proves ineffective, proceeding straight to transplant in this setting would be premature. Interferon rarely has been used since the advent of TKI therapy because of an unfavorable side effect profile.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

A 13-year-old Hispanic girl is found to have an elevated white count of 25,000/mm3 with 80% Auer rod-containing granular blasts that by flow cytometry express very bright CD33 but are negative for human leukocyte antigen- DR isotype. Marrow aspirate shows nearly 100% replacement with blasts. The fluorescence in situ hybridization lab calls you to report that they have evidence of a PML-RARA translocation in the leukemic blasts.>br/>
How do you plan to initiate therapy?
a. Perform a lumbar puncture to determine leukemic involvement because of its high risk in this phenotype, then proceed with induction chemotherapy and all-trans retinoic acid (ATRA) and arsenic trioxide (ATO) therapy followed by bone marrow transplant in first remission.
b. Determine whether coagulopathy is present before obtaining CSF, then start therapy with ATRA, ATO, and chemotherapy on day 1.
c. Start ATRA alone, then begin ATO and chemotherapy days later, followed by bone marrow transplant in first remission.
d. Start induction chemotherapy alone, obtain HLA typing, and start a donor search because of the poor prognosis associated with this leukemic phenotype in association with the high white count.
e. Use ATRA and ATO alone, because the patient’s low white count in this phenotype indicates a good prognosis without the need for conventional chemotherapy.

A

This represents a diagnosis of acute promyelocytic leukemia (APL; M3 in the French-American-British classification), which has an overall favorable prognosis because of its high event-free survival rates with ATRA, arsenic trioxide, and, in high-risk cases, chemotherapy. Thus, stem cell transplant is not indicated in first remission. However, the white count is greater than 10,000/mm3 at diagnosis, suggesting a higher risk APL rather than lower risk. Although ATRA has made dramatic improvements in APL outcome, its use as a single agent sustains remission for only a limited period of time before patients relapse. Until recently, chemotherapy was also thought to be necessary for sustained remissions. Recent data have indicated that prolonged remissions may be possible in lower risk cases with ATRA and arsenic trioxide alone. Differentiation syndrome is a complication of using ATRA alone when the white count is high and should be used concurrently with chemotherapy for patients with initially high white counts. Otherwise, ATRA may be started alone, followed in a few days by traditional chemotherapy. Coagulopathy due to disseminated intravascular coagulation is present in a high percentage of patients with APL, and therefore this should be evaluated before performing a lumbar puncture to avoid the risk of bleeding. Also, central nervous system disease is rare in APL.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

A 1-month-old boy presents with bruising, pallor, poor feeding, and lethargy. He is noted to be tachypneic and hypoxic and has a diffuse interstitial infiltrate on chest X-ray. CBC reveals a WBC count of 650,000/mm3 (95% myeloblasts), hemoglobin of 7 g/dL, and platelet count of 36,000/mm3.
What is the most likely cause of the infiltrate and respiratory symptoms and the most appropriate initial treatment?
a. Hyperleukocytosis; initiation of induction chemotherapy
b. Hyperleukocytosis; leukapheresis or manual exchange transfusion and initiation of induction
chemotherapy
c. Respiratory syncytial virus bronchiolitis; ribavirin d. Mycoplasma pneumonia; azithromycin
e. Reactive airway disease; prednisone and albuterol

A

WBC counts greater than 100,000/mm3 are associated with the clinical syndrome of hyperleukocytosis, especially when the cause of the elevated white count is acute myeloid leukemia. Clinical features of hyperleukocytosis can include CNS findings (eg, lethargy, focal neurologic deficits, intracranial bleeding, hemorrhagic stroke), respiratory findings (tachypnea, dyspnea, hypoxia, and diffuse interstitial infiltrates), and renal dysfunction (often complicated by concomitant tumor lysis syndrome). The pathophysiology of hyperleukocytosis includes increased viscosity of blood and resultant congestion within the capillary beds of the affected organs. Hyperleukocytosis is a medical emergency that requires immediate “debulking” of the circulating tumor burden, which is best accomplished by manual exchange transfusion or by leukapheresis. Initiation of chemotherapy should proceed as soon as possible but should not be the first step in management. The other choices are less likely in the clinical scenario presented.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

An 18-year-old man has been newly diagnosed with acute myeloid leukemia with myelomonocytic characteristics and a marrow blast percentage of 33%. Cytogenetics reveals a 5q deletion. For this patient, what should the optimal therapy include?
a. An anthracycline, intensive cytarabine, and allogeneic hematopoietic stem cell transplant (HSCT)
b. Intensive cytarabine, an anthracycline, and 1 year of maintenance chemotherapy
c. An anthracycline, cyclophosphamide, etoposide, and HSCT
d. Four-drug induction, consolidation, interim maintenance, delayed intensification, and maintenance chemotherapy
e. HSCT without preceding chemotherapy

A

This patient has a diagnosis of acute myeloid leukemia (AML). The backbone of AML therapy includes the intensive administration of anthracyclines and cytarabine without maintenance therapy. Therefore, answers B, C, and D are not appropriate. Optimal therapy for a patient with AML and a high-risk cytogenetic abnormality such as monosomy 7, 5q deletion, or FLT3/ITD high allelic ratio includes remission induction and then allogeneic stem cell transplantation with the best available donor. Proceeding directly to HSCT with 33% blasts in the bone marrow (answer E) would be associated with a very high risk of relapse

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

Fluorescence in situ hybridization testing on the peripheral blood of a 3-month-old boy with newly diagnosed acute leukemia reveals rearrangement of the MLL gene at 11q23. Flow cytometry and morphology are consistent with acute myeloid leukemia (AML) with monocytic differentiation. The patient has an identical twin brother, who is currently asymptomatic, and three older healthy siblings. There is no family history of leukemia or other blood disorders.
What do you tell the family regarding the healthy twin?
a. The probability of the healthy twin developing AML is very low (less than 1%) and is no different from the risk of the older siblings developing AML.
b. The probability of the healthy twin developing AML is about 10%, and if he does develop AML, it will probably take years for the AML to develop.
c. The probability of the healthy twin developing AML is very high (more than 50%), but it will probably take years for the twin to develop AML.
d. The probability of the healthy twin developing AML is very high (more than 50%) and is likely to occur within weeks to months, so the twin should be followed very closely, with frequent (every 1 to 2 weeks) exams and blood work.

A

The concordance rate of leukemia in monozygotic twins is variable depending on the age that the first twin develops leukemia. In this case, the first twin was diagnosed in infancy and, like most infant leukemias, his was characterized by rearrangement of the MLL gene at 11q23. The concordance rate in such cases is very high (and therefore answers A and B are not the best answers), and typically the second twin develops leukemia within weeks to months of the first twin’s diagnosis (and therefore answer C is not the best answer). This indicates that infant leukemias typically initiate in utero (and so the preleukemic clone or fully leukemic clone is shared between the twins because of the common placental circulation that is commonly seen in monozygotic twins) and that preleukemic clones with MLL rearrangements are extremely likely to rapidly progress to full-blown leukemia. Answer B describes a situation in which the first twin was diagnosed in childhood rather than infancy. Answer A describes a situation in which the twins were dizygotic (fraternal), and the placental circulations were separate.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

A 9-year-old boy enters the emergency room with a 1-week history of decreasing strength and numbness in his lower extremities along with midthoracic back pain. He has had no fever, bruising, or history of trauma. Examination reveals decreased strength in the lower extremities, reduced deep tendon reflexes, and downgoing toes. There is slight tenderness in the midthoracic region of his back. An MRI reveals a T6 paraspinal mass with spinal cord compression. A CBC reveals a WBC count of 28,000/mm3 with 23% circulating myeloblasts, a platelet count of 25,000/mm3, and a hemoglobin of 10 g/dL.
Which of the following choices is the most appropriate next step in the evaluation and treatment?
a. Urgent radiation oncology and neurosurgical consultation to address the paraspinal mass, followed by
diagnostic bone marrow aspiration
b. Flow cytometry of peripheral blood followed by appropriate chemotherapy regimen c. Diagnostic bone marrow aspiration followed by radiation to paraspinal mass
d. Flow cytometry of peripheral blood followed by radiation to paraspinal mass
e. Diagnostic bone marrow aspiration followed by appropriate chemotherapy regimen

A

This patient has acute myeloid leukemia (AML) that is complicated by a paraspinal chloroma (solid mass of AML cells). The clinical syndrome of spinal cord compression is a medical emergency that requires immediate local treatment (external beam irradiation or surgery). Steroid treatment is commonly initiated but is not as likely as emergent radiation to help and may cause unnecessary side effects. Thus, steroids should not be considered a substitute for radiation in this setting. The diagnostic procedures are important but must await the treatment of the medical emergency.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

You are evaluating a new test to predict invasive fungal disease in children undergoing chemotherapy for acute myeloid leukemia. You have enrolled 1,000 patients over 7 years. Here are the results:
What is the negative predictive value of the test?
a. 0.25
b. 0.57099999999999995 c. 0.66700000000000004 d. 0.33300000000000002 e. 0.75

A

Explanation
The negative predictive value is the proportion of those who test negative who do not have the disease. In this case, it is 400/600 = 66.7%. A helpful approach here is to create a table of the given information with the screening or diagnostic test results as the rows and the pathological findings as the columns, such as the one below:
Here, TP = true positive; TN = true negative; FP = false positive; and FN = false negative. With the table constructed in this manner, it is straightforward to compute sensitivity, specificity, positive predictive value, and negative predictive value.
Negative predictive value is the probability a subject does not have the disease, given a negative test result. The only portion of the table relevant to this quantity is the second row, representing the screening test negative subjects. TN tells us how many subjects truly did not have the disease among all the negative screening test results, and FN + TN tells us how many subjects yielded a negative screening test. With some simplifying assumptions about the prevalence of the disease, the negative predictive value can be computed as TN/(FN + TN).

31
Q

A 5-year-old boy presents with fulminant acute hepatic failure. He is noted to be bleeding from his gums and nose and has hematochezia. This patient’s bleeding is probably caused by which of the following combinations?
a. Deficiency of fibrinogen, factor VII, and factor II b. Thrombocytopenia and factor XI deficiency
c. Factor VIII, IX, and XI deficiency
d. Factor V and VIII deficiency
e. Low levels of von Willebrand factor and factor VIII

A

Liver failure results in severe derangements in the coagulation system, and although most clotting factors are synthesized in the liver, a number of clotting factors have extrahepatic synthesis either wholly or at least in part. Fibrinogen and factors VII and II are exclusively made in the liver, and thus answer A is correct. Although low platelets are not unusual in patients with liver failure, it is also possible for the platelets to be normal or even elevated as an acute phase reactant. Although factor XI is made in the liver, a combination of thrombocytopenia and factor XI deficiency is not as likely in this scenario as answer A. Although factor VIII is made in the liver, it is also synthesized in extrahepatic sites, and importantly and probably due to being an acute phase reactant, factor VIII levels are often elevated in acute hepatic failure, sometimes significantly. This makes answers C, D, and E incorrect. Regarding answer D, factor V is also synthesized in megakaryocytes and is delivered to the site of bleeding by platelets. Regarding answer E, von Willebrand factor is synthesized in endothelial cells and megakaryocytes, and its production is unaffected by liver disease. Because it is also an acute phase reactant, its levels are often elevated in acute hepatitis, and this is also a reason for an elevated factor VIII in liver disease.

32
Q

A 14-year-old girl with osteomyelitis is receiving antibiotics at home via a percutaneously inserted central catheter (PICC line). She has developed an abscess despite antibiotic therapy and needs incision and drainage. The orthopedic surgeon orders a PT and PTT. The patient has never had any bleeding symptoms. She had 2 teeth extracted when she was 3 years old and a tonsillectomy and adenoidectomy at age 7 years, neither of which resulted in excessive bleeding. She has a PT of 16.2 seconds (normal 9.7 to 11.2 seconds) and a PTT of 61.3 seconds (normal 22 to 36 seconds).
What is the most appropriate next step?
a. Order a fibrinogen level.
b. Order levels of factors II, V, and X.
c. Repeat the PT and PTT.
d. Determine the details of sample procurement.
e. Proceed with the incision and drainage without further testing.

A

This question raises two critical points. First is the need for preoperative laboratory testing, and second is understanding the pitfalls of coagulation testing. With regard to the first point, one could conclude that this child does not have a bleeding disorder based on her negative history for bleeding, which includes two significant hemostatic challenges. Therefore, it would be reasonable to perform this minor procedure without any testing, and one would be tempted to choose answer E; however, given that the tests were already performed, and it is possible for children with bleeding disorders to not bleed excessively with dental extractions and even tonsillectomy and adenoidectomy, it would not be prudent to ignore the test results. Because she has a PICC line, it is possible that the lab tests were drawn from it, and results of coagulation testing from heparinized lines, be they central or peripheral, are not reliable. Therefore, the next most appropriate step is to determine whether the laboratory tests were drawn from the PICC line, and thus the correct answer is D. Ordering factor levels or a fibrinogen level is premature, considering that the abnormalities may be artifactual. Repeating the PT and PTT seems reasonable; however, without knowing where the labs were drawn from, if they were drawn from the PICC line the first time and then repeated from the same place, the results could be the same, which could “strengthen” the argument that she has a bleeding disorder and lead to unnecessary tests or even potentially harmful treatment.

33
Q

Which of the following are the main elements involved in primary hemostasis?
a. Collagen, von Willebrand factor, and platelets b. Factor XII, von Willebrand factor, and platelets c. Factor XI, factor XII, and von Willebrand factor d. Collagen, von Willebrand factor, and fibrinogen e. Platelets, von Willebrand factor, and fibrinogen

A

It is important to understand the various elements of primary hemostasis, which is the first phase of clot formation. When the endothelium breaks and the blood is exposed to the subendothelium, von Willebrand factor binds to the subendothelial collagen and has its platelet binding sites exposed, thus leading to platelet adherence. Thus, answer A is the correct answer. Factors and fibrinogen are part of secondary and not primary hemostasis.

34
Q

Which of the following reasons explains why a patient with factor XII deficiency does not bleed?
a. Activation of factor XI is the key initiator of physiologic coagulation.
b. Factor XII first needs to be activated by pre-kallikrein.
c. Tissue factor and factor VII are the prime initiators of physiologic coagulation. d. Factor XII does not bind to platelets.
e. Factor XII has no role in coagulation.

A

Factor XII is one of the contact factors, and thus its role in initiating coagulation occurs when the blood is exposed to foreign substances such as extracorporeal circuits or central venous catheters, so answer E is incorrect. Factor XII has no role in physiologic coagulation.
In the absence of foreign surfaces, coagulation starts in the subendothelium with activation of tissue factor, which then activates factor VII to factor VIIa, which then activates factor IX and factor X, leading to thrombin generation, making answer C correct. Factor XII is activated in part by pre-kallikrein, but this is not the reason factor XII does not lead to bleeding. Factor XII does not bind to platelets, but this is not why it does not cause bleeding.

35
Q

You are asked to consult on a 12-year-old patient with congenital heart disease and Eisenmenger phenomenon who needs to have a surgical procedure. The surgeon obtained the following pre-op laboratory tests:
PT: 18 seconds (normal 10 to 13 seconds)
PTT: 58 seconds (normal 23 to 36 seconds)
WBC: 18.9 × 109/L
Hgb: 22.3 g/dL
Hct: 68%
Platelet count: 126,000
You decide to order factor levels to determine what factor deficiency the patient may have, but the coagulation laboratory director refuses.
What is the correct next step?
a. Order a mixing study.
b. Repeat the PT and PTT.
c. Repeat the PT and PTT with modifications. d. Order a thrombin time.
e. Order a heparin-neutralized PTT.

A

All coagulation assays drawn into citrate tubes (blue tops) rely on a plasma:citrate ratio of 9:1, and any preanalytic conditions that significantly alter this ratio can affect the results. For example, an underfilled tube will result in a lower ratio of plasma or higher ratio of citrate, which will result in falsely elevated levels due to the citrate effect on the plasma. Similarly, a patient with a very high hematocrit will have, by definition, a lower concentration of plasma, and the effect is the same as that of an underfilled tube-too much citrate for the amount of plasma present, resulting in falsely abnormal results, in this case the PT and PTT. To correct for this, the PT and PTT can be done, but the laboratory must prepare a specialized citrate tube with less citrate so that the ratio of 9:1 plasma to citrate is maintained. Therefore, the correct answer is C.

36
Q

A 10-day-old boy is being seen in the emergency room because of lethargy and poor feeding. His anterior fontanel is full. A CT scan demonstrates an intraparenchymal hemorrhage. Coagulation tests are ordered, with the following results: PT, 37 seconds (normal 9.7 to 11.2 seconds); and PTT, 66 seconds (normal 22 to 36 seconds).
This child may have which of the following factor deficiencies?
a. Factor VII b. Factor VIII c. Factor IX d. Factor X e. Factor XI

A

PT and PTT are screening tests performed to evaluate most of the clotting factors (factor XIII is not evaluated by these assays). It is critical to understand which factors are affected by each assay in order to make the correct diagnosis rapidly, particularly when treatment is warranted as soon as possible. In this scenario, both PT and PTT are prolonged, which means that for a single factor deficiency, that factor would have to reside in the common pathway, which includes fibrinogen and factors II, V, and X. Therefore, the correct answer is D. Factor VII deficiency would not prolong PTT, and deficiencies of factors VIII, IX, or XI would not prolong PT.

37
Q

A 2-week-old infant is referred to the oncology clinic due to the presence of a 10-cm × 5-cm area of swelling on the upper back. On exam, the lesion is firm with a rubbery texture, dark red to purple, and shiny. It is not compromising any function and does not appear to be painful. Of note, the parents also report bruises on the chest and back that are finger-tip shaped and also a bruise over the posterior iliac crest. Which of the following combinations of laboratory tests are most likely to be abnormal for age?
a. von Willebrand factor and factor VIII b. Factors XI and XII
c. Fibrinogen and platelet count
d. Factors II, VII, IX, and X
e. Fibrinogen and factor XIII

A

This patient is presenting at an age and with a presentation consistent with a kaposiform hemangioendothelioma (KHE), and these specific pathologic lesions are associated with Kasabach-Merritt phenomenon, which is a coagulopathy caused by the lesion. The most commonly abnormal findings in Kasabach-Merritt phenomenon are hypofibrinogenemia and thrombocytopenia. Typically, other factor levels are normal for age or are less affected such that the other choices are incorrect. Low von Willebrand factor and FVIII are hallmarks of von Willebrand disease, while low factors II, VII, IX, and X are due to vitamin K deficiency during infancy. Factors XI, XII, and XIII are not excessively consumed by vascular malformations.

38
Q

A 2-week-old infant is referred to the oncology clinic due to the presence of a 10-cm × 5-cm area of swelling on the upper back. On exam, the lesion is firm with a rubbery texture, dark red to purple, and shiny. It is not compromising any function and does not appear to be painful. Of note, the parents also report bruises on the chest and back that are finger-tip shaped and also a bruise over the posterior iliac crest. Other than kaposiform hemangioendothelioma (KHE), the patient is most likely to have which pathology noted on biopsy?
a. Lymphangioma
b. Tufted angioma
c. Angiosarcoma
d. Hemangioblastoma e. Pyogenic granuloma

A

The two lesions most closely associated with Kasabach-Merritt
hemangioendotheliomas (KHE) and tufted angiomas (TA). Lymphangiomas are not associated with coagulopathy. Angiosarcomas are malignant lesions usually found in older patients and not associated with coagulopathy. Hemangioblastomas arise from vessels of the central nervous system and so would not present as skin lesions.
phenomenon are
kaposiform

39
Q

A 2-year-old patient with a hemangioma on her leg since birth is being seen in follow up. The lesion has increased in size three-fold since she was 6 months old. In addition, she is noted to have a number of bruises of different sizes on her shins and forearms. Due to the presence of the hemangioma, a CBC and coagulation testing is done revealing a platelet count of 92 × 109/L (92,000 per dL) and a normal PT and an aPTT of 48 seconds (normal, 25-39 seconds). Which is the best next therapeutic step to manage the abnormal lab tests?
a. Reassurance
b. Start aminocaproic acid
c. Give a platelet transfusion
d. Give a prothrombin complex concentrate e. Give cryoprecipitate

A

This child’s laboratory tests may or may not be related to the hemangioma, but regardless, the bruising seems to be normal toddler bruising based on the location. In the absence of more significant bleeding symptoms, the only “treatment” needed is reassurance. It may be appropriate to evaluate the abnormal laboratory tests further, but this question relates to therapeutic options. The rest of the options are likely to be unhelpful and potential harmful (thrombosis with aminocaproic acid and prothrombin complex concentrates) because of transfusion- related reactions with platelet or cryoprecipitate transfusions.

40
Q

A 3-day-old infant is brought to the emergency department after having a seizure. A CT scan demonstrates massive intracranial hemorrhage. On your examination, the child has numerous bruises on the abdomen and trunk. Which scenario is most likely?
a. The baby was born to a diabetic mother. b. The baby was born at home.
c. The baby is exclusively breastfed.
d. The baby has craniosynostosis.
e. The baby had no prenatal care.

A

This is a classic presentation of vitamin K deficiency bleeding. The so-called classic presentation occurs between 2 and 7 days of age, and babies often present with intracranial hemorrhage. From the above choices, only answer B suggests that this is the diagnosis. Infants born at home are at highest risk for not receiving prophylactic vitamin K at birth. Infants of diabetic mothers are not at risk for bleeding complications, nor are children with craniosynostosis. Exclusively breastfed infants are at risk for late vitamin K deficiency bleeding, which generally occurs at 4 to 12 weeks of age, but they are not at risk for bleeding at this age. A baby with no prenatal care is not necessarily at higher risk for vitamin K deficiency than other babies.

41
Q

A 2-year-old boy with congenital heart disease has a deep vein thrombosis and, after treatment with heparin, develops heparin-induced thrombocytopenia. He is placed on argatroban, a direct thrombin inhibitor. As a result of thrombin inhibition, which of the following effects is expected to occur?
a. Decreased activation of factors VII, IX, and X
b. Increased activation of factor XIII and thrombin-activatable fibrinolysis inhibitor c. Increased conversion of fibrinogen to fibrin and factor V to factor Va
d. Decreased release of von Willebrand factor from Weibel-Palade bodies
e. Decreased activation of factors V and VIII

A

To understand the physiology of the coagulation system, one must understand the function of thrombin. In essence, thrombin’s effects on the coagulation system are all prohemostatic. Specifically, it activates procoagulant factors (V, VIII, XI), activates antifibrinolytic factors (factor XIII, thrombin-activatable fibrinolysis inhibitor [TAFI]), activates platelets, and converts fibrinogen to fibrin. Thus, an inhibitor of thrombin would have the opposite effect, making answer E correct. Answer A is incorrect because thrombin does not activate factors VII, IX, or X, so there would be no effect on those clotting factors. Thrombin activates factor XIII and TAFI, so inhibition of thrombin would do the opposite, making answer B incorrect. Answer C is incorrect for the same reason as answer B, and answer D is incorrect because thrombin is not involved in the release of von Willebrand factor from Weibel-Palade bodies.

42
Q

You are asked to consult on a newborn girl with purpura fulminans. Upon taking the medical history, you learn that this child had a male sibling who died in the neonatal period after presenting with purpura fulminans. She has three other siblings who are healthy and did not have purpura fulminans.
Which physiologic consequence will result from this child’s underlying condition?
a. Excess von Willebrand factor high-molecular-weight multimers b. Decreased fibrinogen
c. Inability to inactivate factor VIII
d. Decreased production of plasminogen
e. Thrombocytopenia

A

The differential diagnosis of neonatal purpura fulminans includes disseminated intravascular coagulation (DIC) and deficiencies of protein C and, less likely, protein S. In this vignette, the child had an older sibling who presented in the same manner, and although DIC can occur with any newborn, this scenario suggests the presence of an autosomal recessive disorder. Therefore, the most likely diagnosis is protein C (or S) deficiency. The protein C/S complex is responsible for inactivating factors V and VIII; thus, the correct answer is C.

43
Q

You are seeing a 12-year-boy old with easy bruising and recurrent epistaxis for a second opinion. He is active in a variety of sports, but his mother thinks that his bruising is excessive. His pediatrician sent the following laboratory tests, all of which are normal: WBC, hemoglobin, platelet count, PT, and PTT. Another hematologist ordered the following, all of which were normal: von Willebrand factor Ag, ristocetin cofactor activity, factor VIII activity, factor XIII activity, and platelet aggregation studies.
Which of the following physical exam findings would be most informative?
a. Petechiae where the blood pressure cuff was placed b. Hypermobility of the finger joints
c. Palpable bruises over the tibial surface
d. A conjunctival hemorrhage
e. Albinism

A

It is not unusual for a hematologist to be referred a patient with bleeding symptoms that are sufficient to warrant concern yet for a detailed laboratory evaluation to be completely negative. Although some of the laboratory testing often warrants repeating (particularly ristocetin cofactor levels), another consideration for such patients is the presence of a connective tissue disorder. This scenario suggests the possibility of Ehlers-Danlos syndrome, a primary collagen disorder in which patients have joint hypermobility, hyperelastic skin, and mucocutaneous bleeding symptoms. Thus, the answer to this question is B. Answers A, C, and D suggest the possibility of a bleeding disorder but would not be informative to the diagnosis. Answer E suggests the possibility of Hermansky- Pudlak syndrome, a disorder characterized by albinism and platelet dysfunction, but in that disorder platelet aggregation studies are abnormal.

44
Q

Patients with a deficiency of plasminogen activator inhibitor type 1 (PAI-1) may have bleeding symptoms for which of the following reasons?
a. They have diminished tissue plasminogen activator (t-PA), leading to bleeding. b. They cannot form a proper fibrin clot.
c. They have decreased ability to activate fibrinogen.
d. They have unopposed effects of t-PA, leading to excess fibrinolysis.
e. The condition is associated with episodes of severe thrombocytopenia.

A

The role of PAI-1, as its name implies, is to inactivate t-PA, and thus a deficiency leads to unopposed t-PA, causing fibrinolytic bleeding, making answer D correct. The other choices are all incorrect characterizations of PAI-1 functions.

45
Q

A 15-year-old girl with cystic fibrosis is going to undergo a partial pneumonectomy due to severe bronchiectasis. The surgeon orders preoperative coagulation testing, which demonstrates a PT of 17.2 seconds (normal 9.7 to 11.2 seconds) and a PTT of 36 seconds (normal 22 to 36 seconds). Because of the abnormality, she is given supplemental oral vitamin K of 5 mg once a day for 3 days over and above the ADEK vitamin she has already been taking. After the third dose, repeat testing demonstrates a PT of 16.9 seconds (normal 9.7 to 11.2 seconds) and a PTT of 37 seconds (normal 22 to 36 seconds). You are asked to consult.
What is the most appropriate next step?
a. Increase the oral vitamin K dosage and repeat the testing.
b. Perform a mixing study on the PT.
c. Give a parenteral dose of vitamin K and repeat the testing.
d. Proceed with surgery with a preoperative dose of recombinant factor VIIa.
e. Proceed with surgery with a preoperative dose of a prothrombin complex concentrate.

A

Vitamin K is a fat-soluble vitamin that can be poorly absorbed in patients with fat malabsorption disorders. In cystic fibrosis, pancreatic dysfunction leads to malabsorption of fat-soluble vitamins, and such patients received ADEK (an oral fat-soluble supplement with vitamins A, D, E, and K). However, patients with cystic fibrosis are at risk for vitamin K deficiency. In this scenario, the patient received a trial of oral vitamin K supplementation with no benefit. The dosage this patient received was adequate, and increasing the dosage is unlikely to have an effect. A mixing study is not likely to yield useful information because only the PT is prolonged. Lupus anticoagulants affect PTT much more commonly than PT, and this clinical scenario is not suggestive for the presence of a lupus anticoagulant. Giving recombinant factor VIIa or prothrombin complex concentrate is likely to correct the abnormality, but they are very expensive medications that carry the risk for thrombosis, and giving them in the absence of a diagnosis is not appropriate. Repeating the vitamin K challenge with a parenteral dose of vitamin K could be both diagnostic and therapeutic. If the repeat testing is normal, then the diagnosis of vitamin K deficiency is confirmed, and in fact the patient has been treated. If the repeat testing is abnormal, then more tests would be indicated. Of note, if such a patient were not to proceed immediately to surgery (eg, if it were to be weeks later), then repeating the PT and PTT would be important because an additional dose of parenteral vitamin K might be needed.

46
Q

A 7-year-old girl with Diamond-Blackfan anemia undergoes stem cell transplantation from her 2-year-old HLA- identical brother. At day 100 neutrophils and platelets are fully reconstituted; however, the patient remains red cell transfusion dependent. The marrow reveals red cell aplasia. Genetic analysis reveals 100% donor status. Of the following, what is the most likely cause of this failure of erythroid engraftment?
a. Anti-red cell antibodies
b. Donor with Diamond-Blackfan mutations c. Graft-versus-host disease (GVHD)
d. Inadequate conditioning
e. Inadequate cell dose

A

In matched sibling donor transplants for all forms of inherited bone marrow failure syndromes (IBMFS), it is essential to critically evaluate the donor for evidence of the same IBMFS. Differences in clinical expression of these disorders may lead to siblings with identical genetic constitutional abnormalities with completely normal or very abnormal blood counts. This requires that the donor be genetically tested for the mutation present in the recipient (if known). In Diamond-Blackfan anemia, evaluation of an erythrocyte adenosine deaminase (eADA) also may be helpful because mutations are only known for approximately 60-75% of clinically diagnosed patients. Anti- red cell antibodies should lead to anemia and hemolysis not failure of erythroid engraftment. GVHD, inadequate conditioning or cell dose could lead to failure to engraft all cell lines, but should not lead to selective erythroid non-engraftment particularly with a marrow that is 100% donor. The most likely explanation is that the donor also carried the same disease-causing Diamond-Blackfan anemia mutations as the patient, despite the presumably normal pre-transplant evaluation including blood counts.

47
Q

A 15-year-old female adolescent returns for routine follow up 8 years after immunosuppressive therapy (IST) with anti-thymocyte globulin and cyclosporine for severe aplastic anemia. She had a good response to IST with normalization of her counts. Annual PNH clone testing has been shown no loss of GPI-linked proteins. She still does not have any siblings. Her blood counts unexpectedly show pancytopenia with a hemoglobin 8.5 g/dL, MCV 108 fL, WBC of 1,200/μL with 10% neutrophils, and platelets of 15,000/μL. What is the most important next step in management?
a. Analyze DNA breakage analysis with DEB/MMC b. Bone marrow evaluation with cytogenetics
c. Measure PNH clone size by flow cytometry
d. Obtain HLA typing
e. Perform telomere length testing

A

Relapse of aplastic anemia following immunosuppressive therapy (IST) with anti-human thymocyte globulin (ATG) and cyclosporine is common (occurring in up to 30% of patients in some series), but relapse occurring this late is unusual. Patients treated with IST are at increased risk for developing clonal cytogenetic abnormalities, including monosomy 7, as well as leukemic transformation. Thus, a bone marrow aspirate and biopsy with cytogenetics would be the next step to evaluate the etiology of her recurrent pancytopenia and guide treatment. If morphologic evidence of myelodysplastic syndrome, cytogenetic abnormalities, or leukemia are present, therapy would include hematopoietic stem cell transplantation (HSCT).
Although reevaluation at relapse should include reassessment of the size of the population of glycosylphosphatidylinositol (GPI)-deficient cells (paroxysmal nocturnal hemoglobinuria [PNH] clone), this is not the most important test to be evaluated. Diepoxybutane (DEB) or mitomycin C (MMC) testing for DNA breakage should have been done with the first presentation, and a patient with Fanconi anemia or another breakage syndrome would have been unlikely to respond to immunosuppressive therapy and maintained normal counts for years. Patients who experience a relapse of their aplastic anemia soon after immunosuppression is withdrawn may be rescued with cyclosporine alone or with combined IST. HLA typing for an unrelated donor should have been done at initial presentation, even without a sibling to be prepared for a poor response to IST. Telomere length testing may be of interest, but again it may have been done with initial presentation and does not influence the care of a patient who had a long initial response to IST. At this time, the most important diagnostic evaluation is to reexamine the bone marrow with morphology and cytogenetics to rule out the acquisition of malignant or premalignant changes.

48
Q

An 8-month-old, gastrostomy-tube dependent, ex-premature infant is referred with WBC of 5,000/mm3, ANC 650/mm3, hemoglobin 10.8 g/dL, mean corpuscular volume (MCV) 100 fL, and platelet count of 200,000/mm3/L. Prothrombin time (PT) is slightly prolonged, but partial thromboplastin time (PTT) and fibrinogen are normal. Serum B12 and folate, liver enzymes, and bilirubin levels are normal. She has chronic smelly diarrhea and is growing slowly despite adequate caloric intake. There is no history of fever or infections either in the stool or systemically. A sweat chloride test for cystic fibrosis and workup for celiac disease are negative.
A mutation in which gene might account for the constellation of symptoms in this child?
a. c-Mpl b. ELA2 c. RPS19 d. SAMD9 e. SBDS

A

This child has moderate neutropenia without a history of infection, mild anemia with macrocytosis but normal B12 and folate, and a normal platelet count. While this could all be reactive to the gastrointestinal process and a consequence of prematurity, failure to thrive, steatorrhea, and the elevated PT most likely related to vitamin K deficiency are consistent with fat malabsorption. The combination of exocrine pancreatic insufficiency and otherwise idiopathic neutropenia is strongly suggestive of Shwachman-Diamond syndrome, associated with mutations in the SBDS gene in the vast majority of cases. Mutations in c-Mpl are associated with congenital amegakaryocytic thrombocytopenia and clinical thrombocytopenia, which is not present here. ELA2 mutations are associated with severe congenital neutropenia (SCN) or cyclic neutropenia (CN), and chronic diarrhea due to exocrine pancreatic insufficiency is not characteristic of either SCN or CN. RPS19 mutations are associated with Diamond-Blackfan anemia, in which neutropenia may occur, but this disorder generally presents with macrocytic red cell aplasia without malabsorption secondary to pancreatic insufficiency. SAMD9 mutations are associated with immunodeficiency and adrenal insufficiency rather than pancreatic issues.

49
Q

A 4-year-old girl with a previously normal CBC now presents in your office with a hemoglobin of 8.5 g/dL, corrected reticulocyte count of 0.1%, and mean corpuscular volume of 80 fl. White cells and platelets are normal in number and morphology. Bilirubin, LDH, BUN, creatinine, and urinalysis are normal. Direct and indirect antiglobulin tests are negative. Workup for infection, including parvovirus, is negative. Occult blood in her stools is negative. Physical examination is unremarkable. She has had no restriction in her energy or activities and the family agrees she is “fine.”
What is the most appropriate next step in management?
a. Administer erythropoietin.
b. Initiate a red cell transfusion.
c. Observe serial hemoglobin values closely. d. Prescribe oral iron supplement.
e. Send red cell adenosine deaminase (eADA).

A

This patient has normocytic red cell aplasia, the most common causes of which are Diamond-Blackfan anemia (DBA) and transient erythroblastopenia of childhood (TEC). DBA, which is often associated with an elevated erythrocyte adenosine deaminase level, typically presents in infancy with macrocytic red cell aplasia but may present in adulthood with normalization of the mean corpuscular volume (MCV) for age. TEC typically presents at an older age than DBA, and the MCV is typically normal. In an asymptomatic patient, transfusion may be deferred at this hemoglobin level. Thus, this patient is most likely to have TEC, and close clinical observation of her blood counts (and clinical status) for the need for a transfusion is the indicated management. An eADA may help to clarify your thinking that this is TEC, but likely, by the time you have the result, blood counts will have recovered and it will be unnecessary. Close observation will still be required. Erythropoietin levels are typically already high with red cell aplasia, and additional exogenous dosing is not beneficial. Although she is anemic, the MCV does not demonstrate microcytosis, which should predate the anemia if the cause were iron insufficient hematopoiesis.

50
Q

Name the main 4 main Genotypes of SCD

A

Hemoglobin SS
Hemoglobin SC
Hemoglobin Sβ Thal0
Hemoglobin Sβ Thal+

51
Q

What is the mechanism of Hydroxyurea

A

HU is ribonucleotide reductase inhibitor
Increases the expression of the gamma globin loci
Hb tetramers containing gamma globin inhibit polymerization of Sickle hemoglobin, reducing hemolysis and increasing Hb level

52
Q

18 year old male with SCD with VOC is on IV morphine and IV ketorolac with uncontrolled pain, What agent can be added?

A

IV Ketamine

53
Q

What is the mechanism of Crizanlizumab?

A

Monoclonal antibody that inhibits P- Selectin on the surface of platelets and endothelium in the blood vessels and has been shown to inhibit interactions between endothelial cells, platelets, red blood cells, sickled red blood cells, and leukocytes, causing a blockade

Weekly IV infusion, Makes RBC, WBC and plt less sticky reducing VOC for patients 16 and above. Younger patient in clinical trials

54
Q

What is the best way to minimize risk of alloimmunization in SCD patients?

A

Performing extended typing of blood antigen as part of routine care.

55
Q

Which factor deficiency does not affect PT or PTT?

A

Factor XIII

56
Q

A family with known severe factor X deficiency has recently moved to your clinic from Pakistan. Patient had ICH at birth. What treatment options can you offer?

A

Prophylaxis with Factor X replacement product (Coagadex)
What is there is no access to factor X replacement products?
PCC: II, VII, IX and X
Cryo: factor VIII, XIII and fibrinogen
FFP: all clotting factors including factor II, VII, VIII, IX, X and XI, vWD, protein c and S (but requires large volume to correct specific factor deficiency)

57
Q

6 year old with recurrent weekly epistaxis lasting 30 mins. Normal CBC, PT, PTTLabs: Factor VIII activity 15%
Von Willebrand antigen: 40%
Von Willebrand activity: 40%
What is the next diagnostic test?

A

Von Willebrand Factor-2N binding assay

58
Q

3 year old male with mild thrombocytopenia, epistaxis and bruising.
Smear shows large platelets without leukocyte inclusions
- abnormal platelet aggregation study. What is the diagnosis?

A

Bernard-Soulier Syndrome

Glanzmann thrombasthenia is a congenital bleeding disorder caused by a deficiency of the platelet integrin alpha IIb beta3. This integrin is the platelet fibrinogen receptor and is thus essential to platelet aggregation and hemostasis.

59
Q

What test is needed to diagnose Dyskeratosis Congenita?

A

Telomere Length testing

Triad: Reticulated skin hyperpigmentation, dystrophic nails, mucosal membrane leukoplakia with age
Aplastic anemia up to 50% by 20-30s
Solid organ cancer, leukemia by 30-40s

Other things: Early graying and pulmonary disease

Most common mutation 35% is x linked DKC1

60
Q

2 month neonate with macrocytic anemia and reticulocytopenia,required two transfusions. Prior to performing a bone marrow evaluation, what other diagnostic test can be helpful?

A

eADA (Erythrocyte adenosine deaminase activity)
In Diamond-Blackfan anemia patients which present with macrocytic pure red cell aplasia, Elevated eADA level can be seen with DBA

61
Q

What is the diagnostic criteria for severe aplastic anemia

A

Bone marrow cellularity less than 25%
Two of the following
ANC <500
ARC <40,000
Platelet count<20,000

Or bone marrow cellularity 25-50% but with less than 30% hematopoietic cells

62
Q

16 year old with dyskeratosis congenita now has rapid progressive decline in blood counts & transfusion dependence. He is an only child, what is the next best treatment option?

A

Unrelated matched donor transplant with reduced intensity conditioning

63
Q

9 year old adopted AA male with pancytopenia was diagnosed with Fanconi anemia. He does not have a matched donor. What treatment options can you offer?

A

Androgen treatment
(Danazol or Oxymetholon)

64
Q

4 year old boy with thrombocytopenia, eczema and maternal uncle with lymphoma.
Smear shows small platelets
What is the diagnosis?

A

Wiskott-Aldrich Syndrome

65
Q

4 year old boy with refractory autoimmune hemolytic anemia. Now has swollen lymph nodes for 6 months +, splenomegaly. CBC shows anemia and thrombocytopenia, Lymphocyte subset is normal. IG G is elevated. What is the next best test?

A

TCR ⍺β CD4- and CD8- T cells
Autoimmune lymphoproliferative syndrome - ALPS

66
Q

Multiple cutaneous hemangioma are usually associated with internal hemangiomas, what common organ should be assessed next?

A

liver
Always obtain an US of the liver if >5 cutaneous hemangiomas
If you have diffuse liver hemangiomas then must check thyroid, because it’s associated with hypothyroidism

67
Q

3 day old infant with Kaposiform Hemangioendothelioma. Name the phenomenon with profound thrombocytopenia, hypofibrinogenemia and high D dimer in this patient

Tends to involve the trunk, usually present at birth (can appear post natally)

Involvement of the skin (redish purple, tense appearance , subcutaneous fat and mucle is typical
Ill defined borders

A

Kasabach-Merritt Phenomenon

68
Q

What blood bank process is needed to make CMV safe blood products

A

Leukoreduction

69
Q

16 year old F with extensive left leg DVT without any known provoking factors, no family history, thrombophilia workup negative .Must evaluate the patient for this syndrome?

A

May-Thurner Syndrome
Narrowed left iliac vein due to chronic compression from right iliac artery

70
Q

congenital or acquired absence/decrease of the von Willebrand factor-cleaving protease ADAMTS13 (a disintegrin and metalloproteinase with a thrombospondin type 1 motif member 13). Low levels of ADAMTS13 activity result in microthrombi formation, which leads to end-organ ischemia and damage.

A

Thrombotic Thrombocytopenic purpura

71
Q

This is a congenital immune deficiency with warts, hypogammaglobulinemia, infections, myelokathexis.Common presenting finding can be neutropenia.

A

WHIM Syndrome (CXCR4 gene)

72
Q

What is the mechanism by which hepcidin regulates iron hemostasis?

A

Hepcidin inhibits iron release from macrophages
Hepcidin is a small peptide made in the liver that appears to have an anti inflammatory function. Its production is induced by IL 6. Its role in iron metabolism is to block iron transport from intestinal mucosal cells and macrophages by binding to and degrading ferroportin. IT is the key mediator in the pathogenesis of anemia of inflammation

73
Q

he most common nondeletional α-thalassemic mutation and is an important cause of HbH-like disease in Southeast Asia.
genotype (αα/ααCS), and have a thalassaemia trait phenotype. The blood film features are identical to that of HbH disease

A

Hemoglobin Constant spring

74
Q
A