Spring 2022 1
SPRING 2022
Leader
in Transition
Dr. Lucio Gordan
assumes new,
‘data-driven’ role
2 FCS Magazine
In the phase 3 RESPONSE* trial, Jakafi demonstrated superior results† vs BAT‡
(25/110) of patients receiving Jakafi achieved Hct control and ≥35% spleen volume
reduction at week 32 vs <1% (1/112) of patients receiving BAT (P < 0.0001)11§
23%
Composite Primary Endpoint
For adults with polycythemia vera (PV) who have had an inadequate response to or are intolerant of hydroxyurea (HU)
Indications and Usage
Jakafi is indicated for treatment of polycythemia vera (PV) in adults who
have had an inadequate response to or are intolerant of hydroxyurea.
Important Safety Information
• Treatment with Jakafi® (ruxolitinib) can cause thrombocytopenia,
anemia and neutropenia, which are each dose-related effects. Perform
a pre-treatment complete blood count (CBC) and monitor CBCs every 2
to 4 weeks until doses are stabilized, and then as clinically indicated
• Manage thrombocytopenia by reducing the dose or temporarily
interrupting Jakafi. Platelet transfusions may be necessary
• Patients developing anemia may require blood transfusions and/or dose
modifications of Jakafi
• Severe neutropenia (ANC <0.5 × 109/L) was generally reversible by
withholding Jakafi until recovery
• Serious bacterial, mycobacterial, fungal and viral infections have
occurred. Delay starting Jakafi until active serious infections have
resolved. Observe patients receiving Jakafi for signs and symptoms of
infection and manage promptly. Use active surveillance and
prophylactic antibiotics according to clinical guidelines
• Tuberculosis (TB) infection has been reported. Observe patients taking
Jakafi for signs and symptoms of active TB and manage promptly. Prior
to initiating Jakafi, evaluate patients for TB risk factors and test those
at higher risk for latent infection. Consult a physician with expertise in
the treatment of TB before starting Jakafi in patients with evidence of
active or latent TB. Continuation of Jakafi during treatment of active TB
should be based on the overall risk-benefit determination
• Progressive multifocal leukoencephalopathy (PML) has occurred with
Jakafi treatment. If PML is suspected, stop Jakafi and evaluate
• Advise patients about early signs and symptoms of herpes zoster and to
seek early treatment
• Increases in hepatitis B viral load with or without associated elevations in
alanine aminotransferase and aspartate aminotransferase have been
reported in patients with chronic hepatitis B virus (HBV) infections. Monitor
and treat patients with chronic HBV infection according to clinical guidelines
• When discontinuing Jakafi, myeloproliferative neoplasm-related
symptoms may return within one week. After discontinuation, some
patients with myelofibrosis have experienced fever, respiratory
distress, hypotension, DIC, or multi-organ failure. If any of these occur
after discontinuation or while tapering Jakafi, evaluate and treat any
intercurrent illness and consider restarting or increasing the dose of
Jakafi. Instruct patients not to interrupt or discontinue Jakafi without
consulting their physician. When discontinuing or interrupting Jakafi for
reasons other than thrombocytopenia or neutropenia, consider gradual
tapering rather than abrupt discontinuation
• Non-melanoma skin cancers (NMSC) including basal cell, squamous cell, and
Merkel cell carcinoma have occurred. Perform periodic skin examinations
• Treatment with Jakafi has been associated with increases in total
cholesterol, low-density lipoprotein cholesterol, and triglycerides. Assess
lipid parameters 8-12 weeks after initiating Jakafi. Monitor and treat
according to clinical guidelines for the management of hyperlipidemia
• Another JAK-inhibitor has increased the risk of major adverse
cardiovascular events (MACE), including cardiovascular death, myocardial
infarction, and stroke (compared to those treated with tumor TNF
In a subset of patients, these characteristics may
indicate advanced PV despite treatment with HU
at the maximum tolerated dose and phlebotomy.1,7-10
Hct
≥45%
WBC count
>11 × 109/L
Disease-related
SYMPTOMS
or
BAT, best available therapy; Hct, hematocrit; MPN-SAF, Myeloproliferative Neoplasm Symptom Assessment Form; TSS, Total Symptom Score; WBC, white blood cell.
*The RESPONSE (Randomized study of Efficacy and Safety in POlycythemia vera with JAK iNhibitor ruxolitinib verSus bEst available care) trial was a randomized, open-label, active-controlled phase
3 trial comparing Jakafi with BAT in 222 patients with PV. Patients enrolled in the study had been diagnosed with PV for at least 24 weeks, had an inadequate response to or were intolerant of HU,
required phlebotomy for Hct control, and exhibited splenomegaly. All patients were required to demonstrate Hct control between 40% and 45% prior to randomization. After week 32, patients were
able to cross over to Jakafi treatment.11,12
†The composite primary endpoint was defined as Hct control without phlebotomy eligibility and a ≥35% spleen volume reduction as measured by CT or MRI. To achieve the Hct control endpoint, patients
could not become eligible for phlebotomy between weeks 8 and 32. Phlebotomy eligibility was defined as Hct >45% that is ≥3 percentage points higher than baseline or Hct >48% (lower value).11,12
‡BAT included HU (60%), interferon/pegylated interferon (12%), anagrelide (7%), pipobroman (2%), lenalidomide/thalidomide (5%), and observation (15%).11
§Jakafi 95% CI, 0.15-0.32; BAT 95% CI, 0.00-0.05.11
In a subset of patients, these characteristics may
indicate advanced PV despite treatment with HU
at the maximum tolerated dose and phlebotomy.1,7-10
PV is a hematologic malignancy that can
become advanced in a subset of patients1-6
adults with polycythemia vera (PV) who ha
V)
PV is a hematologic malignancy t
become advanced in a subset of
EVERY DAY COUNTS.
JAKAFI CAN HELP.
Spring 2022 3
aPatients with data at both baseline (value >0) and week 32 were included in this analysis. Negative values indicate a reduction in the
severity of symptoms.12
RESPONSE was an open-label trial and, therefore, not designed to evaluate differences in symptoms11
Patient-reported outcomes were assessed using the MPN-SAF symptom diary. The MPN-SAF diary was administered daily in an
electronic diary format to score 14 disease-related symptoms on a scale of 0 (absent) to 10 (worst possible). At baseline, median TSS
was 23.4 (range, 0-106) in the group receiving Jakafi and 33.3 (range, 0-118) in the group receiving BAT.12,15
Exploratory Endpoint
0.4%
BAT
Jakafi
40
20
-20
-40
-60
-80
-100
Median Change in Symptom Score
From Baseline to Week 32, %
-49.6%
-94.9%
-61.1%
-99.5% -100%
-51.5%
-44%
-80.2%
-64.1%
-41.8%
0%
-37.1%
-93.9%
-65.9%
-4.2%
-2.1%
3.9%
-4.4%
11.1%
16.7%
7.9%
5%
10.9%
17.2%
15.7%
0%
1.4%
Cytokine
Symptoms
Hyperviscosity
Symptoms
Splenomegaly
Symptoms
Tiredness
Itching
Muscle Ache
Sweating While Awake
Night Sweats
Concentration Problems
Headache
Dizziness
Vision Problems
Ringing in Ears
Numbness or Tingling
in Hands or Feet
Skin Redness
Early Satiety
Abdominal Discomfort
Median Percent Change in Symptom Score From Baseline to Week 3212a
At week 32, 49% (36/74) of patients receiving Jakafi and
5% (4/81) of patients receiving BAT had at least a 50%
reduction in the 14-item MPN-SAF TSS12
Patients receiving Jakafi had reductions in all symptom
clusters reported, whereas patients receiving BAT had
an increase in scores of many symptoms12
From The New England Journal of Medicine, Vannucchi AM, Kiladjian JJ,
Griesshammer M, et al, Ruxolitinib versus Standard Therapy for the Treatment of
Polycythemia Vera, 372(5), 426-435. Copyright © 2015 Massachusetts Medical
Society. Reprinted with permission from Massachusetts Medical Society.
To achieve the Hct control endpoint, patients could not become eligible for phlebotomy between weeks 8 and 32. Phlebotomy eligibility was defined as Hct >45%
that is ≥3 percentage points higher than baseline or Hct >48% (lower value)11,12
(66/110) of patients receiving Jakafi achieved Hct control at week 32 vs 19% (21/112) of patients receiving BAT11
60%
Individual Component of the Primary Endpoint
Analysis was conducted in week 32 Hct control
responders, beginning at week 3213
Progression events for the evaluation of
duration of absence of phlebotomy eligibility
included fi rst of 2 consecutive Hct assessments
that confi rms phlebotomy eligibility, death, or
development of MF or acute leukemia15
Reprinted from The Lancet Haematology, 7(3), Kiladjian J-J, Zachee P,
Hino M, Long-term efficacy and safety of ruxolitinib versus best available
therapy in polycythaemia vera (RESPONSE): 5-year follow up of a phase
3 study, e226-e237, Copyright 2020, with permission from Elsevier.
100
80
60
40
20
12
24
36
48
60
72
84
96
108
120
132
144
156
168
180
192
204
216
228
240
252
264
Probability, %
No. of responders/events/censor: 66/16/50
Duration of Response, wk
+ Censoring times
– Jakafi
No. at risk
Events
66 66 66 66 66 66 64 63 62 60 58 58 56 56 56 56 56 55 54 54 53 51 49 49 48 48 48 44 44 44 42 42 42 41 40 39 39 39 39 38 28 5 3 1 0
0 0 0 0 0 0 0 1 2 4 5 5 6 6 6 6 6 7 8 8 9 10 10 10 10 10 10 12 12 12 14 14 14 15 15 16 16 16 16 16 16 16 16 16 16
Probability of Maintaining Hct Controla at 5 Years in RESPONSE Trial13,14
aAbsence of phlebotomy eligibility
73%
Kaplan-Meier Estimate: Durability of Hct Control at 5 Years
(95% CI, 0.60-0.83)
Intervene with Jakafi in your appropriate patients with advanced PV
INTERVENEJAKAFI.COM
INTERVENE WITH JAKAFI IN YOUR PATIENTS WITH ADVANCED PV
blockers) in patients with rheumatoid arthritis, a condition for which Jakafi
is not indicated. Consider the benefits and risks for the individual patient
prior to initiating or continuing therapy with Jakafi particularly in patients
who are current or past smokers and patients with other cardiovascular risk
factors. Patients should be informed about the symptoms of serious
cardiovascular events and the steps to take if they occur
• Another JAK-inhibitor has increased the risk of thrombosis, including
deep venous thrombosis (DVT), pulmonary embolism (PE), and arterial
thrombosis (compared to those treated with TNF blockers) in patients
with rheumatoid arthritis, a condition for which Jakafi is not indicated.
In patients with myelofibrosis (MF) and polycythemia vera (PV) treated
with Jakafi in clinical trials, the rates of thromboembolic events were
similar in Jakafi and control treated patients. Patients with symptoms of
thrombosis should be promptly evaluated and treated appropriately
• Another JAK-inhibitor has increased the risk of lymphoma and other
malignancies excluding NMSC (compared to those treated with TNF
blockers) in patients with rheumatoid arthritis, a condition for which
Jakafi is not indicated. Patients who are current or past smokers are at
additional increased risk. Consider the benefits and risks for the
individual patient prior to initiating or continuing therapy with Jakafi,
particularly in patients with a known secondary malignancy (other than
a successfully treated NMSC), patients who develop a malignancy, and
patients who are current or past smokers
• In myelofibrosis and polycythemia vera, the most common nonhematologic
adverse reactions (incidence ≥15%) were bruising, dizziness, headache,
and diarrhea. In acute graft-versus-host disease, the most common
nonhematologic adverse reactions (incidence >50%) were infections
(pathogen not specified) and edema. In chronic graft-versus-host disease,
the most common nonhematologic adverse reactions (incidence ≥20%)
were infections (pathogen not specified) and viral infections
• Avoid concomitant use with fluconazole doses greater than 200 mg.
Dose modifications may be required when administering Jakafi with
fluconazole doses of 200 mg or less, or with strong CYP3A4 inhibitors,
or in patients with renal or hepatic impairment. Patients should be
closely monitored and the dose titrated based on safety and efficacy
• Use of Jakafi during pregnancy is not recommended and should only be used if
the potential benefit justifies the potential risk to the fetus. Women taking Jakafi
should not breastfeed during treatment and for 2 weeks after the final dose
Please see Brief Summary of Full Prescribing Information for Jakafi on
the following pages. To learn more about Jakafi, visit HCP.Jakafi.com
References: 1. Barosi G, et al. Br J Haematol. 2010;148(6):961-963.
2. Parasuraman S, et al. Exp Hematol Oncol. 2016;5:3. 3. Mascarenhas J. Clin
Lymphoma Myeloma Leuk. 2016;16 Suppl:S124-S129. 4. Rumi E, et al. Blood.
2017;129(6):680-692. 5. Michiels JJ, et al. World J Hematol. 2013;2(3):71-88.
6. Michiels JJ. World J Crit Care Med. 2015;4(3):230-239. 7. Marchioli R, et al.
N Engl J Med. 2013;368(1):22-33. 8. Barbui T, et al. Blood. 2015;126(4):560-561.
9. Emanuel RM, et al. J Clin Oncol. 2012;30(33):4098-4103. 10. Verstovsek S, et al.
Cancer. 2014;120(4):513-520. 11. Jakafi Prescribing Information. Wilmington, DE:
Incyte Corporation. 12. Vannucchi AM, et al. N Engl J Med. 2015;372(5):426-435.
13. Kiladjian J-J, et al. Lancet Haematol. 2020;7(3):e226-e237. 14. Kiladjian J-J, et al.
Lancet Haematol. 2020;7(Suppl):1-18. 15. Data on file. Incyte Corporation. Wilmington, DE.
Jakafi and the Jakafi logo are registered trademarks of Incyte.
© 2022, Incyte Corporation. MAT-JAK-03757 01/22
BRIEF SUMMARY: For Full Prescribing Information,
see package insert.
INDICATIONS AND USAGE Myelofibrosis Jakafi is
indicated for treatment of intermediate or high-risk
myelofibrosis (MF), including primary MF,
post-polycythemia vera MF and post-essential
thrombocythemia MF in adults. Polycythemia Vera Jakafi
is indicated for treatment of polycythemia vera (PV) in
adults who have had an inadequate response to or are
intolerant of hydroxyurea. Acute Graft-Versus-Host
Disease Jakafi is indicated for treatment of steroid-
refractory acute graft-versus-host disease (aGVHD) in adult
and pediatric patients 12 years and older. Chronic Graft-
Versus-Host Disease Jakafi is indicated for treatment of
chronic graft-versus-host disease (cGVHD) after failure of
one or two lines of systemic therapy in adult and pediatric
patients 12 years and older.
CONTRAINDICATIONS None.
WARNINGS AND PRECAUTIONS Thrombocytopenia,
Anemia and Neutropenia Treatment with Jakafi can
cause thrombocytopenia, anemia and neutropenia. [see
Adverse Reactions (6.1) in Full Prescribing Information].
Manage thrombocytopenia by reducing the dose or
temporarily interrupting Jakafi. Platelet transfusions may
be necessary [see Dosage and Administration (2) in Full
Prescribing Information]. Patients developing anemia may
require blood transfusions and/or dose modifications of
Jakafi. Severe neutropenia (ANC less than 0.5 × 109/L)
was generally reversible by withholding Jakafi until
recovery. Perform a pre-treatment complete blood count
(CBC) and monitor CBCs every 2 to 4 weeks until doses
are stabilized, and then as clinically indicated [see
Dosage and Administration (2) in Full Prescribing
Information]. Risk of Infection Serious bacterial,
mycobacterial, fungal and viral infections have occurred
[see Adverse Reactions (6.1) in Full Prescribing
Information]. Delay starting therapy with Jakafi until
active serious infections have resolved. Observe patients
receiving Jakafi for signs and symptoms of infection and
manage promptly. Use active surveillance and
prophylactic antibiotics according to clinical guidelines.
Tuberculosis Tuberculosis infection has been reported in
patients receiving Jakafi. Observe patients receiving
Jakafi for signs and symptoms of active tuberculosis and
manage promptly. Prior to initiating Jakafi, patients
should be evaluated for tuberculosis risk factors, and
those at higher risk should be tested for latent infection.
Risk factors include, but are not limited to, prior residence
in or travel to countries with a high prevalence of
tuberculosis, close contact with a person with active
tuberculosis, and a history of active or latent tuberculosis
where an adequate course of treatment cannot be
confirmed. For patients with evidence of active or latent
tuberculosis, consult a physician with expertise in the
treatment of tuberculosis before starting Jakafi. The
decision to continue Jakafi during treatment of active
tuberculosis should be based on the overall risk-benefit
determination. Progressive Multifocal
Leukoencephalopathy Progressive multifocal
leukoencephalopathy (PML) has occurred with Jakafi
treatment. If PML is suspected, stop Jakafi and evaluate.
Herpes Zoster Advise patients about early signs and
symptoms of herpes zoster and to seek treatment as
early as possible if suspected. Hepatitis B Hepatitis B viral
load (HBV-DNA titer) increases, with or without associated
elevations in alanine aminotransferase and aspartate
aminotransferase, have been reported in patients with
chronic HBV infections taking Jakafi. The effect of Jakafi
on viral replication in patients with chronic HBV infection
is unknown. Patients with chronic HBV infection should
be treated and monitored according to clinical guidelines.
Symptom Exacerbation Following Interruption or
Discontinuation of Treatment with Jakafi Following
discontinuation of Jakafi, symptoms from
myeloproliferative neoplasms may return to pretreatment
levels over a period of approximately one week. Some
patients with MF have experienced one or more of the
following adverse events after discontinuing Jakafi: fever,
respiratory distress, hypotension, DIC, or multi-organ
failure. If one or more of these occur after discontinuation
of, or while tapering the dose of Jakafi, evaluate for and
treat any intercurrent illness and consider restarting or
increasing the dose of Jakafi. Instruct patients not to
interrupt or discontinue Jakafi therapy without consulting
their physician. When discontinuing or interrupting
therapy with Jakafi for reasons other than
thrombocytopenia or neutropenia [see Dosage and
Administration (2.7) in Full Prescribing Information],
consider tapering the dose of Jakafi gradually rather than
discontinuing abruptly. Non-Melanoma Skin Cancer
(NMSC) Non-melanoma skin cancers including basal cell,
squamous cell, and Merkel cell carcinoma have occurred
in patients treated with Jakafi. Perform periodic skin
examinations. Lipid Elevations Treatment with Jakafi has
been associated with increases in lipid parameters
including total cholesterol, low-density lipoprotein (LDL)
cholesterol, and triglycerides [see Adverse Reactions (6.1)
in Full Prescribing Information]. The effect of these lipid
parameter elevations on cardiovascular morbidity and
mortality has not been determined in patients treated
with Jakafi. Assess lipid parameters approximately 8-12
weeks following initiation of Jakafi therapy. Monitor and
treat according to clinical guidelines for the management
of hyperlipidemia. Major Adverse Cardiovascular
Events (MACE) Another JAK-inhibitor has increased the
risk of MACE, including cardiovascular death, myocardial
infarction, and stroke (compared to those treated with
TNF blockers) in patients with rheumatoid arthritis, a
condition for which Jakafi is not indicated. Consider the
benefits and risks for the individual patient prior to
initiating or continuing therapy with Jakafi particularly in
patients who are current or past smokers and patients
with other cardiovascular risk factors. Patients should be
informed about the symptoms of serious cardiovascular
events and the steps to take if they occur. Thrombosis
Another JAK-inhibitor has increased the risk of
thrombosis, including deep venous thrombosis (DVT),
pulmonary embolism (PE), and arterial thrombosis
(compared to those treated with TNF blockers) in patients
with rheumatoid arthritis, a condition for which Jakafi is
not indicated. In patients with MF and PV treated with
Jakafi in clinical trials, the rates of thromboembolic
events were similar in Jakafi and control treated patients.
Patients with symptoms of thrombosis should be
promptly evaluated and treated appropriately. Secondary
Malignancies Another JAK-inhibitor has increased the
risk of lymphoma and other malignancies excluding
NMSC (compared to those treated with TNF blockers) in
patients with rheumatoid arthritis, a condition for which
Jakafi is not indicated. Patients who are current or past
smokers are at additional increased risk. Consider the
benefits and risks for the individual patient prior to
initiating or continuing therapy with Jakafi, particularly in
patients with a known secondary malignancy (other than
a successfully treated NMSC), patients who develop a
malignancy, and patients who are current or past
smokers. ADVERSE REACTIONS The following clinically
significant adverse reactions are discussed in greater
detail in other sections of the labeling: • Thrombocytopenia,
Anemia and Neutropenia [see Warnings and Precautions
(5.1) in Full Prescribing Information] • Risk of Infection
[see Warnings and Precautions (5.2) in Full Prescribing
Information] • Symptom Exacerbation Following
Interruption or Discontinuation of Treatment with Jakafi
[see Warnings and Precautions (5.3) in Full Prescribing
Information ] • Non-Melanoma Skin Cancer [see Warnings
and Precautions (5.4) in Full Prescribing Information]
• Lipid Elevations [see Warnings and Precautions (5.5)
in Full Prescribing Information] • Major Adverse
Cardiovascular Events (MACE) [see Warnings and Precautions
(5.6) in Full Prescribing Information] • Thrombosis [see
Warnings and Precautions (5.7) in Full Prescribing
Information] • Secondary Malignancies [see Warnings and
Precautions (5.8) in Full Prescribing Information]. Clinical
Trials Experience Because clinical trials are conducted
under widely varying conditions, adverse reaction rates
observed in the clinical trials of a drug cannot be directly
compared to rates in the clinical trials of another drug
and may not reflect the rates observed in practice.
Myelofibrosis The safety of Jakafi was assessed in 617
patients in six clinical studies with a median duration of
follow-up of 10.9 months, including 301 patients with MF in
two Phase 3 studies. In these two Phase 3 studies,
patients had a median duration of exposure to Jakafi of
9.5 months (range 0.5 to 17 months), with 89%
of patients treated for more than 6 months and 25% treated
for more than 12 months. One hundred and eleven (111)
patients started treatment at 15 mg twice daily and 190
patients started at 20 mg twice daily. In patients starting
treatment with 15 mg twice daily (pretreatment platelet
counts of 100 to 200 × 109/L) and 20 mg twice daily
(pretreatment platelet counts greater than 200 × 109/L),
65% and 25% of patients, respectively, required a dose
reduction below the starting dose within the first 8 weeks
of therapy. In a double-blind, randomized, placebo-
controlled study of Jakafi, among the 155 patients
treated with Jakafi, the most frequent adverse reactions
were thrombocytopenia and anemia [see Table 2].
Thrombocytopenia, anemia and neutropenia are dose-related
effects. The three most frequent nonhematologic adverse
reactions were bruising, dizziness and headache [see
Table 1]. Discontinuation for adverse events, regardless of
causality, was observed in 11% of patients treated with Jakafi
and 11% of patients treated with placebo. Table 1 presents
the most common nonhematologic adverse reactions
occurring in patients who received Jakafi in the double-blind,
placebo-controlled study during randomized treatment.
Table 1: Myelofibrosis: Nonhematologic Adverse
Reactions Occurring in Patients on Jakafi in
the Double-blind, Placebo-controlled Study
During Randomized Treatment
Jakafi
(N=155)
Placebo
(N=151)
Adverse
Reactions
All
Gradesa
(%)
Grade
3
(%)
Grade
4
(%)
All
Grades
(%)
Grade
3
(%)
Grade
4
(%)
Bruisingb
23
< 1
15
Dizzinessc
18
< 1
Headache
15
Urinary Tract
Infectionsd
< 1
< 1
Weight Gaine
< 1
< 1
Flatulence
< 1
Herpes Zosterf
< 1
a National Cancer Institute Common Terminology Criteria for Adverse Events
(CTCAE), version 3.0
b includes contusion, ecchymosis, hematoma, injection site hematoma,
periorbital hematoma, vessel puncture site hematoma, increased tendency
to bruise, petechiae, purpura
c includes dizziness, postural dizziness, vertigo, balance disorder, Meniere’s
Disease, labyrinthitis
d includes urinary tract infection, cystitis, urosepsis, urinary tract infection
bacterial, kidney infection, pyuria, bacteria urine, bacteria urine identified,
nitrite urine present
e includes weight increased, abnormal weight gain
f includes herpes zoster and post-herpetic neuralgia
Description of Selected Adverse Reactions: Anemia
In the two Phase 3 clinical studies, median time to
onset of first CTCAE Grade 2 or higher anemia was
approximately 6 weeks. One patient (< 1%) discontinued
treatment because of anemia. In patients receiving Jakafi,
mean decreases in hemoglobin reached a nadir of
approximately 1.5 to 2.0 g/dL below baseline after 8 to 12
weeks of therapy and then gradually recovered to reach a
new steady state that was approximately 1.0 g/dL below
baseline. This pattern was observed in patients regardless
of whether they had received transfusions during therapy.
In the randomized, placebo-controlled study, 60% of
patients treated with Jakafi and 38% of patients receiving
placebo received red blood cell transfusions during
randomized treatment. Among transfused patients, the
median number of units transfused per month was 1.2
in patients treated with Jakafi and 1.7 in placebo treated
patients. Thrombocytopenia In the two Phase 3 clinical
studies, in patients who developed Grade 3 or 4
thrombocytopenia, the median time to onset was
approximately 8 weeks. Thrombocytopenia was generally
reversible with dose reduction or dose interruption.
The median time to recovery of platelet counts above
50 × 109/L was 14 days. Platelet transfusions were
administered to 5% of patients receiving Jakafi and to 4%
of patients receiving control regimens. Discontinuation
Spring 2022 5
of treatment because of thrombocytopenia occurred in
< 1% of patients receiving Jakafi and < 1% of patients
receiving control regimens. Patients with a platelet count
of 100 × 109/L to 200 × 109/L before starting Jakafi had
a higher frequency of Grade 3 or 4 thrombocytopenia
compared to patients with a platelet count greater than
200 × 109/L (17% versus 7%). Neutropenia In the two
Phase 3 clinical studies, 1% of patients reduced or stopped
Jakafi because of neutropenia. Table 2 provides the
frequency and severity of clinical hematology abnormalities
reported for patients receiving treatment with Jakafi or
placebo in the placebo-controlled study.
Table 2: Myelofibrosis: Worst Hematology Laboratory
Abnormalities in the Placebo-Controlled Studya
Jakafi
(N=155)
Placebo
(N=151)
Laboratory
Parameter
All
Gradesb
(%)
Grade
3
(%)
Grade
4
(%)
All
Grades
(%)
Grade
3
(%)
Grade
4
(%)
Thrombocytopenia
70
31
Anemia
96
34
11
87
16
Neutropenia
19
< 1
a Presented values are worst Grade values regardless of baseline
b National Cancer Institute Common Terminology Criteria for Adverse Events,
version 3.0
Additional Data from the Placebo-Controlled Study
• 25% of patients treated with Jakafi and 7% of patients
treated with placebo developed newly occurring or
worsening Grade 1 abnormalities in alanine transaminase
(ALT). The incidence of greater than or equal to Grade 2
elevations was 2% for Jakafi with 1% Grade 3 and no
Grade 4 ALT elevations. • 17% of patients treated with
Jakafi and 6% of patients treated with placebo developed
newly occurring or worsening Grade 1 abnormalities in
aspartate transaminase (AST). The incidence of Grade 2
AST elevations was < 1% for Jakafi with no Grade 3 or 4
AST elevations. • 17% of patients treated with Jakafi and
< 1% of patients treated with placebo developed newly
occurring or worsening Grade 1 elevations in cholesterol.
The incidence of Grade 2 cholesterol elevations was
< 1% for Jakafi with no Grade 3 or 4 cholesterol
elevations. Polycythemia Vera In a randomized,
open-label, active-controlled study, 110 patients with PV
resistant to or intolerant of hydroxyurea received Jakafi
and 111 patients received best available therapy [see
Clinical Studies (14.2) in Full Prescribing Information].
The most frequent adverse reaction was anemia.
Discontinuation for adverse events, regardless of
causality, was observed in 4% of patients treated with
Jakafi. Table 3 presents the most frequent nonhematologic
adverse reactions occurring up to Week 32.
Table 3: Polycythemia Vera: Nonhematologic Adverse
Reactions Occurring in ≥ 5% of Patients on
Jakafi in the Open-Label, Active-controlled
Study up to Week 32 of Randomized Treatment
Jakafi
(N=110)
Best Available
Therapy (N=111)
Adverse Reactions
All
Gradesa
(%)
Grade
3-4
(%)
All
Grades
(%)
Grade
3-4
(%)
Diarrhea
15
< 1
Dizzinessb
15
13
Dyspneac
13
Muscle Spasms
12
< 1
Constipation
Herpes Zosterd
< 1
Nausea
Weight Gaine
< 1
Urinary Tract Infectionsf
Hypertension
< 1
< 1
a National Cancer Institute Common Terminology Criteria for Adverse Events
(CTCAE), version 3.0
b includes dizziness and vertigo
c includes dyspnea and dyspnea exertional
d includes herpes zoster and post-herpetic neuralgia
e includes weight increased and abnormal weight gain
f includes urinary tract infection and cystitis
Clinically relevant laboratory abnormalities are shown
in Table 4.
Table 4: Polycythemia Vera: Selected Laboratory
Abnormalities in the Open-Label, Active-
controlled Study up to Week 32 of
Randomized Treatmenta
Jakafi
(N=110)
Best Available
Therapy (N=111)
Laboratory
Parameter
All
Gradesb
(%)
Grade
3
(%)
Grade
4
(%)
All
Grades
(%)
Grade
3
(%)
Grade
4
(%)
Hematology
Anemia
72
< 1
< 1
58
Thrombocytopenia
27
< 1
24
< 1
Neutropenia
< 1
10
< 1
Chemistry
Hypercholesterolemia
35
Elevated ALT
25
< 1
16
Elevated AST
23
23
< 1
Hypertriglyceridemia
15
13
a Presented values are worst Grade values regardless of baseline
b National Cancer Institute Common Terminology Criteria for Adverse Events,
version 3.0
Acute Graft-Versus-Host Disease In a single-arm,
open-label study, 71 adults (ages 18-73 years) were
treated with Jakafi for aGVHD failing treatment with
steroids with or without other immunosuppressive drugs
[see Clinical Studies (14.3) in Full Prescribing Information].
The median duration of treatment with Jakafi was 46 days
(range, 4-382 days). There were no fatal adverse reactions
to Jakafi. An adverse reaction resulting in treatment
discontinuation occurred in 31% of patients. The most
common adverse reaction leading to treatment
discontinuation was infection (10%). Table 5 shows the
adverse reactions other than laboratory abnormalities.
Table 5: Acute Graft-Versus-Host Disease:
Nonhematologic Adverse Reactions Occurring
in ≥ 15% of Patients in the Open-Label, Single-
Cohort Study
Jakafi (N=71)
Adverse Reactionsa
All Gradesb (%)
Grade 3-4 (%)
Infections (pathogen
not specified)
55
41
Edema
51
13
Hemorrhage
49
20
Fatigue
37
14
Bacterial infections
32
28
Dyspnea
32
Viral infections
31
14
Thrombosis
25
11
Diarrhea
24
Rash
23
Headache
21
Hypertension
20
13
Dizziness
16
a Selected laboratory abnormalities are listed in Table 6 below
b National Cancer Institute Common Terminology Criteria for Adverse Events
(CTCAE), version 4.03
Selected laboratory abnormalities during treatment with
Jakafi are shown in Table 6.
Table 6: Acute Graft-Versus-Host Disease: Selected
Laboratory Abnormalities Worsening from
Baseline in the Open-Label, Single Cohort Study
Jakafi (N=71)
Worst grade during treatment
Laboratory Parameter
All Gradesa (%)
Grade 3-4 (%)
Hematology
Anemia
75
45
Thrombocytopenia
75
61
Neutropenia
58
40
Chemistry
Elevated ALT
48
Elevated AST
48
Hypertriglyceridemia
11
a National Cancer Institute Common Terminology Criteria for Adverse Events,
version 4.03
Chronic Graft-Versus-Host Disease In a Phase 3,
randomized, open-label, multi-center study, 165 patients
were treated with Jakafi and 158 patients were treated
with best available therapy for cGVHD failing treatment
with steroids with or without other immunosuppressive
drugs [see Clinical Studies (14.4) in full Prescribing
Information]; sixty-five patients crossed over from best
available therapy to treatment with Jakafi, for a total of
230 patients treated with Jakafi. The median duration of
exposure to Jakafi for the study was 49.7 weeks (range, 0.7
to 144.9 weeks) in the Jakafi arm. One hundred and nine
(47%) patients were on Jakafi for at least 1 year. There were
five fatal adverse reactions to Jakafi, including 1 from toxic
epidermal necrolysis and 4 from neutropenia, anemia and/or
thrombocytopenia. An adverse reaction resulting in treatment
discontinuation occurred in 18% of patients treated with
Jakafi. An adverse reaction resulting in dose modification
occurred in 27%, and an adverse reaction resulting in
treatment interruption occurred in 23%. The most common
hematologic adverse reactions (incidence > 35%) are
anemia and thrombocytopenia. The most common
nonhematologic adverse reactions (incidence ≥ 20%) are
infections (pathogen not specified) and viral infection. Table 7
presents the most frequent nonlaboratory adverse reactions
occurring up to Cycle 7 Day 1 of randomized treatment.
Table 7: Chronic Graft-Versus-Host Disease: All-Grade
(≥ 10%) and Grades 3-5 (≥ 3%) Nonlaboratory
Adverse Reactions Occurring in Patients in the
Open-Label, Active-controlled Study up to Cycle
7 Day 1 of Randomized Treatment
Jakafi
(N = 165)
Best Available
Therapy (N = 158)
Adverse Reactionsb
All
Gradesa
(%)
Grade
≥ 3
(%)
All
Grades
(%)
Grade
≥ 3
(%)
Infections and infestations
Infections (pathogen
not specified)
45
15
44
16
Viral infections
28
23
Musculoskeletal and connective tissue disorders
Musculoskeletal pain
18
13
General disorders and administration site conditions
Pyrexia
16
2
9
1
Fatigue
13
1
10
2
Edema
10
1
12
1
Vascular disorders
Hypertension
16
5
13
7
Hemorrhage
12
2
15
2
Respiratory, thoracic and mediastinal disorders
Cough
13
0
8
0
Dyspnea
11
1
8
1
Gastrointestinal disorders
Nausea
12
0
13
2
Diarrhea
10
1
13
1
a National Cancer Institute Common Terminology Criteria for Adverse Events
(CTCAE), version 4.03
b Grouped terms that are composites of applicable adverse reaction terms.
Clinically relevant laboratory abnormalities are shown in
Table 8.
Table 8: Chronic Graft-Versus-Host Disease: Selected
Laboratory Abnormalities in the Open-Label,
Active-controlled Study up to Cycle 7 Day 1
of Randomized Treatmenta
Jakafi
(N=165)
Best Available
Therapy (N=158)
Laboratory Test
All
Gradesb
(%)
Grade
≥ 3
(%)
All
Grades
(%)
Grade
≥ 3
(%)
Hematology
Anemia
82
13
75
8
Thrombocytopenia
27
12
23
9
Neutropenia
58
20
54
17
Chemistry
Hypercholesterolemia
88
10
85
Elevated AST
65
54
Elevated ALT
73
11
71
16
Gamma
glutamyltransferase
increased
81
42
75
38
Creatinine increased
47
40
Elevated lipase
38
12
30
Elevated amylase
35
25
a Presented values are worst Grade values regardless of baseline
b National Cancer Institute Common Terminology Criteria for Adverse Events,
version 4.03
DRUG INTERACTIONS Fluconazole Concomitant use
of Jakafi with fluconazole increases ruxolitinib exposure
[see Clinical Pharmacology (12.3) in Full Prescribing
Information], which may increase the risk of exposure-
related adverse reactions. Avoid concomitant use of
Jakafi with fluconazole doses of greater than 200 mg
daily. Reduce the Jakafi dosage when used concomitantly
with fluconazole doses of less than or equal to 200 mg
[see Dosage and Administration (2.5) in Full Prescribing
Information]. Strong CYP3A4 Inhibitors Concomitant
use of Jakafi with strong CYP3A4 inhibitors increases
ruxolitinib exposure [see Clinical Pharmacology (12.3) in
Full Prescribing Information], which may increase the risk
of exposure-related adverse reactions. Reduce the Jakafi
dosage when used concomitantly with strong CYP3A4
inhibitors except in patients with aGVHD or cGVHD
[see Dosage and Administration (2.5) in Full Prescribing
Information]. Strong CYP3A4 Inducers Concomitant use
of Jakafi with strong CYP3A4 inducers may decrease
ruxolitinib exposure [see Clinical Pharmacology (12.3) in
Full Prescribing Information], which may reduce efficacy
of Jakafi. Monitor patients frequently and adjust the
Jakafi dose based on safety and efficacy [see Clinical
Pharmacology (12.3) in Full Prescribing Information].
USE IN SPECIFIC POPULATIONS Pregnancy: Risk
Summary When pregnant rats and rabbits were
administered ruxolitinib during the period of
organogenesis adverse developmental outcomes
occurred at doses associated with maternal toxicity
(see Data). There are no studies with the use of Jakafi
in pregnant women to inform drug-associated risks. The
background risk of major birth defects and miscarriage
for the indicated populations is unknown. Adverse
outcomes in pregnancy occur regardless of the health
of the mother or the use of medications. The background
risk in the U.S. general population of major birth defects
is 2% to 4% and miscarriage is 15% to 20% of clinically
recognized pregnancies. Data: Animal Data Ruxolitinib
was administered orally to pregnant rats or rabbits during
the period of organogenesis, at doses of 15, 30 or
60 mg/kg/day in rats and 10, 30 or 60 mg/kg/day in
rabbits. There were no treatment-related malformations.
Adverse developmental outcomes, such as decreases of
approximately 9% in fetal weights were noted in rats at
the highest and maternally toxic dose of 60 mg/kg/day.
This dose results in an exposure (AUC) that is
approximately 2 times the clinical exposure at the
maximum recommended dose of 25 mg twice daily.
In rabbits, lower fetal weights of approximately 8%
and increased late resorptions were noted at the highest
and maternally toxic dose of 60 mg/kg/day. This dose
is approximately 7% the clinical exposure at the
maximum recommended dose. In a pre- and post-natal
development study in rats, pregnant animals were dosed
with ruxolitinib from implantation through lactation at
doses up to 30 mg/kg/day. There were no drug-related
adverse findings in pups for fertility indices or for
maternal or embryofetal survival, growth and
development parameters at the highest dose evaluated
(34% the clinical exposure at the maximum
recommended dose of 25 mg twice daily). Lactation:
Risk Summary No data are available regarding the
presence of ruxolitinib in human milk, the effects on
the breast fed child, or the effects on milk production.
Ruxolitinib and/or its metabolites were present in the milk
of lactating rats (see Data). Because many drugs are
present in human milk and because of the potential for
thrombocytopenia and anemia shown for Jakafi in human
studies, discontinue breastfeeding during treatment with
Jakafi and for two weeks after the final dose. Data:
Animal Data Lactating rats were administered a single
dose of [14C]-labeled ruxolitinib (30 mg/kg) on postnatal
Day 10, after which plasma and milk samples were
collected for up to 24 hours. The AUC for total
radioactivity in milk was approximately 13-fold the
maternal plasma AUC. Additional analysis showed the
presence of ruxolitinib and several of its metabolites in
milk, all at levels higher than those in maternal plasma.
Pediatric Use The safety and effectiveness of Jakafi for
treatment of myelofibrosis or polycythemia vera in
pediatric patients have not been established. The safety
and effectiveness of Jakafi for treatment of
Jakafi is a registered trademark of Incyte.
U.S. Patent Nos. 7598257; 8415362; 8722693; 8822481;
8829013; 9079912; 9814722; 10016429
© 2011-2021 Incyte Corporation.
Revised: September 2021 PLR-JAK-00054
steroid-refractory aGVHD has been established for
treatment of children 12 years and older. Use of Jakafi
in pediatric patients with steroid-refractory aGVHD
is supported by evidence from adequate and
well-controlled trials of Jakafi in adults [see Clinical
Studies (14.3) in Full Prescribing Information] and
additional pharmacokinetic and safety data in pediatric
patients. The safety and effectiveness of Jakafi for
treatment of steroid-refractory aGVHD has not been
established in pediatric patients younger than 12 years
old. The safety and effectiveness of Jakafi for treatment
of cGVHD after failure of one or two lines of systemic
therapy has been established for treatment of children
12 years and older. Use of Jakafi in pediatric patients
with cGVHD after failure of one or two lines of systemic
therapy is supported by evidence from adequate and
well-controlled trials of Jakafi in adults and adolescents
[see Clinical Studies (14.3, 14.4) in Full Prescribing
Information] and additional pharmacokinetic and safety
data in pediatric patients. The safety and effectiveness of
Jakafi for treatment of cGVHD has not been established in
pediatric patients younger than 12 years old. Jakafi was
evaluated in a single-arm, dose-escalation study
(NCT01164163) in 27 pediatric patients with relapsed or
refractory solid tumors (Cohort A) and 20 with leukemias or
myeloproliferative neoplasms (Cohort B). The patients had
a median age of 14 years (range, 2 to 21 years) and
included 18 children (age 2 to < 12 years), and 14
adolescents (age 12 to < 17 years). The dose levels tested
were 15, 21, 29, 39, or 50 mg/m2 twice daily in 28-day
cycles with up to 6 patients per dose group. Overall, 38
(81%) patients were treated with no more than a single
cycle of Jakafi, while 3, 1, 2, and 3 patients received 2, 3, 4,
and 5 or more cycles, respectively. A protocol-defined
maximal tolerated dose was not observed, but since few
patients were treated for multiple cycles, tolerability with
continued use was not assessed adequately to establish a
recommended Phase 2 dose higher than the recommended
dose for adults. The safety profile in children was similar
to that seen in adults. Juvenile Animal Toxicity Data
Administration of ruxolitinib to juvenile rats resulted in
effects on growth and bone measures. When administered
starting at postnatal day 7 (the equivalent of a human
newborn) at doses of 1.5 to 75 mg/kg/day, evidence of
fractures occurred at doses ≥ 30 mg/kg/day, and effects
on body weight and other bone measures [e.g., bone
mineral content, peripheral quantitative computed
tomography, and x-ray analysis] occurred at doses
≥ 5 mg/kg/day. When administered starting at postnatal
day 21 (the equivalent of a human 2-3 years of age) at
doses of 5 to 60 mg/kg/day, effects on body weight and
bone occurred at doses ≥ 15 mg/kg/day, which were
considered adverse at 60 mg/kg/day. Males were more
severely affected than females in all age groups, and
effects were generally more severe when administration
was initiated earlier in the postnatal period. These
findings were observed at exposures that are at least
27% the clinical exposure at the maximum recommended
dose of 25 mg twice daily. Geriatric Use Of the total
number of patients with MF in clinical studies with Jakafi,
52% were 65 years and older, while 15% were 75 years
and older. No overall differences in safety or effectiveness
of Jakafi were observed between these patients and
younger patients. Clinical studies of Jakafi in patients
with aGVHD did not include sufficient numbers of subjects
age 65 and over to determine whether they respond
differently from younger subjects. Of the total number of
patients with cGVHD treated with Jakafi in clinical trials,
11% were 65 years and older. No overall differences in
safety or effectiveness of Jakafi were observed between
these patients and younger patients. Renal Impairment
Total exposure of ruxolitinib and its active metabolites
increased with moderate (CLcr 30 to 59 mL/min) and
severe (CLcr 15 to 29 mL/min) renal impairment, and
ESRD (CLcr less than 15 mL/min) on dialysis [see Clinical
Pharmacology (12.3) in Full Prescribing Information].
Modify Jakafi dosage as recommended [see Dosage and
Administration (2.6) in full Prescribing Information].
Hepatic Impairment Exposure of ruxolitinib increased
with mild (Child-Pugh A), moderate (Child-Pugh B) and
severe (Child-Pugh C) hepatic impairment [see Clinical
Pharmacology (12.3) in full Prescribing Information].
Reduce Jakafi dosage as recommended in patients with
MF or PV with hepatic impairment [see Dosage and
Administration (2.6) in full Prescribing Information].
Reduce Jakafi dosage as recommended for patients with
Stage 4 liver aGVHD. Monitor blood counts more
frequently for toxicity and modify the Jakafi dosage for
adverse reactions if they occur for patients with Score 3
liver cGVHD [see Dosage and Administration (2.6) and
Clinical Pharmacology (12.3) in full Prescribing
Information]. OVERDOSAGE There is no known antidote
for overdoses with Jakafi. Single doses up to 200 mg
have been given with acceptable acute tolerability. Higher
than recommended repeat doses are associated with
increased myelosuppression including leukopenia,
anemia and thrombocytopenia. Appropriate supportive
treatment should be given. Hemodialysis is not expected
to enhance the elimination of Jakafi.
Spring 2022 7
DEPARTMENTS
26
People and Places
29
Foundation Update
30
From Our Patients
SPOTLIGHTS
16
Data Governance
18
Strategic Partnership
20
Value-Based Care
22
FCS Operations Team Meeting
FEATURES
10
Physician Leaders Take on
New Roles in the New Year
14
Conquering Breast Cancer
IN
THIS
ISSUE
10
22
18
14
We welcome your feedback, article suggestions
and photos (high resolution please).
Email to FCSCommunications@FLCancer.com
On the cover: Dr. Lucio Gordan with one of his
horses at his Gainesville farm.
Photography by Blake Jones
© Annexus Health, Inc. 2022. All Rights Reserved. AH013 4/22
We have a lot in
common with you—
The patient is at the heart
of everything we do
Scan the QR code below with your smartphone or visit
annexushealth.com/fcs to view a personal message
from our Co-Founder and CEO Joe Baffone to all of you
at Florida Cancer Specialists. We are passionate about
helping you help your patients gain access to the care
they need—faster and with less financial toxicity.
Spring 2022 9
PHYSICIAN LEADERSHIP
PRESIDENT & MANAGING PHYSICIAN
MICHAEL DIAZ, MD
EXECUTIVE LEADERSHIP
CHIEF EXECUTIVE OFFICER
NATHAN H. WALCKER
CHIEF OPERATING OFFICER
JASON COE
CHIEF ADMINISTRATIVE OFFICER
JOYCE NELSON
CHIEF FINANCIAL OFFICER
RICH MACCLARY
CHIEF INFORMATION OFFICER
KEN STURTZ
CHIEF LEGAL OFFICER & GENERAL COUNSEL
NANCY ARDELL
CHIEF PROCUREMENT OFFICER
PAUL CHADWICK
CHIEF COMPLIANCE OFFICER
VALERIE EASTWOOD
PUBLISHED BY
IN PARTNERSHIP WITH
NATHAN H. WALCKER,
CHIEF EXECUTIVE OFFICER
Across our practice, we have
embraced this new year of
opportunities and challenges at
full speed.
Data increasingly drives the
actions we take and the decisions we
make. In this issue, we shine the spotlight on our recently
launched Data Governance Program. It’s a significant
initiative that is enhancing our ability to consistently
provide exceptional patient care.
You’ll also want to read about our value-based care
activities, which continue to rank FCS among the top-
performing oncology practices in the country for quality
and cost-saving measures.
At all times our work is centered around those who
entrust their care to us. There is no greater reward than to
hear from our patients that choosing FCS was their best
decision ever.
Amy Rodriquez, a breast cancer survivor, tells of her
experience with Dr. David Wright and the Tampa Cancer
Center team. She describes how every person she
encountered welcomed and embraced her like family. The
care and support she received enabled her to return to
military service, cancer free.
I could not be more proud of the hard work and
dedication of our talented and caring physicians and team
members. Together we will continue to push FCS to new
heights of success and deliver on our promise to our patients
by providing world-class cancer care, close to home.
MICHAEL DIAZ, MD,
PRESIDENT & MANAGING
PHYSICIAN
As an oncologist, it is a privilege
and an amazing opportunity to
help make the world a better place
for cancer patients. In my new
role as FCS President & Managing
Physician, I look forward to helping FCS achieve many
more milestones as an innovative leader in community-
based oncology care.
While I am energized by the successes that are ahead
of us, certain things will remain constant — above all, our
commitment as one team to one mission.
We will continue to lead the way in cutting-edge cancer
innovations and discoveries and introduce supportive
services that ease stress for patients and families and
improve their overall quality of life.
Another constant is our steadfast commitment to
improving the access and affordability of cancer care.
No voices are better equipped to make an impact
than our dedicated CPAN advocates. They are the
patients, survivors, caregivers and clinicians who have
experienced cancer firsthand. We are so fortunate
for their partnership, and in this issue, you will learn
how they are helping eliminate barriers to life-saving
community-based cancer care.
Every day, it is our privilege and our passion to serve
our patients. Thank you for all you do to help us deliver
on our mission.
10 FCS Magazine
LEADERSHIP FEATURE
Spring 2022 11
LEADERSHIP FEATURE
BY EMMA WITMER, PHOTOS BY BLAKE JONES
A
bit of rearranging at the executive level has positioned
FCS to achieve new heights of success.
Michael Diaz, MD, FCS Executive Board member,
Director of Patient Advocacy and medical oncologist at
two FCS locations in St. Petersburg, began the new year as FCS
President & Managing Physician. He succeeds Lucio Gordan, MD,
who, after serving as FCS’s physician leader for three years,
has stepped into a new role that will allow him to continue
supporting strategic initiatives in clinical advancements, value-
based care and much more.
“Our journey in creating our national reputation
has involved many years of hard work, dedication and
commitment, and we are so grateful to Dr. Gordan for his
outstanding leadership,” said FCS Chief Executive Officer
Nathan H. Walcker. “Dr. Diaz is perfectly positioned to
take on this role at this time,” he added, citing his long-term
commitment to patients and his career focus on ensuring
access and affordability to cancer care.
Diaz has been influential in both state and national policy,
often organizing grassroots campaigns to block legislation
that could pose harm to cancer patients. As a member of
the Florida Medicaid Pharmaceutical and Therapeutics
Committee, Diaz has worked to revamp the practices of
pharmacy benefit managers and helped develop alternative
oncology payment models with the American Society of
Clinical Oncology, Community Oncology Alliance and the
Centers for Medicare and Medicaid Services Innovation.
Diaz says, “Patients need and deserve to get excellent quality,
and there is no reason that should be sacrificed for monetary
reasons. You can balance both, and that’s what I have been
striving to do.”
As FCS President & Managing Physician, Diaz is
committed to working on behalf of patients and healthcare
Michael Diaz, MD
President & Managing
Physician
Lucio Gordan, MD
Chief Medical Officer of
Therapeutics and Analytics
Physician Leaders
Take on New Roles
in the New Year
Following his passion,
Chief Medical Officer
of Therapeutics and
Analytics Dr. Lucio
Gordan shifts his focus
to data and technology
and spending more
time with his family, wife
Valeria and daughter
Julia, on their farm.
12 FCS Magazine
providers and continuing his focus on
quality, research, advocacy and innovation.
His priorities involve networking with
other medical disciplines to centralize
patient care, addressing health disparities
with pharmaceutical companies and
investing in more personalized treatment
through the continued advancement of
next-generation sequencing.
“It’s an amazing honor and privilege
to take on the role of president,” Diaz
said. “My colleagues have expressed their
confidence in what I’ve done in the past,
and they are trusting me to continue to do
that in the future. I take that very seriously.
I am going to continue to do everything I
can to earn that confidence and respect.”
For Gordan, his new role as Chief
Medical Officer of Therapeutics and
Analytics is a thrilling opportunity to
dive more deeply into a topic that has
been a lifelong passion and can prove
revolutionary in the quality of care offered
to FCS patients.
Data and informatics have always
fascinated Gordan. As a child growing up
in Brazil, Gordan taught himself to do
coding and developed computer software
beginning at age 11. He was insatiably
curious about how data could be used to
help others and to improve understanding
of the challenges in the world around him.
As a practicing medical oncologist with
FCS’s Gainesville Cancer Center, Gordan
understands the importance of data to
improve patient care.
Data allows clinicians to provide
personalized treatment based on key
factors in the patient’s demographic
information, medical history and
genomics. A data-driven approach
expedites the process of finding the right
medications and leads to improvement
in drug adherence, dose density and dose
intensity. Well-organized data is also a
key factor in getting qualified patients
into potentially life-saving clinical trials.
FCS is sitting on a treasure trove of data.
It’s a massive undertaking to sort through
all of it, but with the help of the Data
Governance Committee and other FCS
teams, Gordan is ready for the challenge.
“As it was during my time as President
& Managing Physician, my goal as Chief
Medical Officer of Therapeutics and
Analytics is to move the needle a bit faster
to impact our full patient population,”
he said.
Spring 2022 13
LEADERSHIP FEATURE
Gordan reserves weekend mornings for exercising
his horses. He owns a 30-acre farm near Gainesville,
where he keeps a few mini-donkeys and a dozen
horses. His favorite horse, a Hanoverian-Tennessee
Walker named Redford, is always up for a ride.
“They are my psychiatrists on staff,” Gordan
said of his horses, with whom he spends regular
non-office hours. “When I was president, the
executives knew not to call me between 8 a.m.
and noon on Saturday and Sunday. If they did,
they knew not to expect an answer.”
Refreshed by pastoral pursuits, Gordan
returns to data, whose utility he understands
and appreciates.
A DIFFERENT KIND OF SADDLE AND REINS PROVIDE PASTIME AND PASSION
14 FCS Magazine
hose who know Amy Rodriguez will
tell you she’s more than a fighter.
She’s a warrior.
Her 17 years of service as
a U.S. Army National Guard Staff
Sergeant have earned Rodriguez more
than a dozen honors, including the Joint
Service Commendation Medal, the Army
Achievement Medal and the Army
Commendation Medal.
Still, Rodriguez’s recent triumph over
breast cancer is her sweetest victory yet.
After a long and arduous year-and-a-half
of treatment, Rodriguez is grateful for the
chance to keep serving her community and
country and to continue raising her son
and watching him grow up.
“I have FCS to thank for that,”
Rodriguez said.
The Tampa native received her breast
cancer diagnosis in 2020, after a concerning
self-exam.
“I had an aunt who had breast cancer
years prior, so I was very adamant about
regularly checking myself,” she said. “When
I found a lump, I didn’t think much of it,
but I mentioned it to my mother-in-law.
She insisted I get it checked out.”
Two weeks later, Dr. Abigail Beard
at USF Health confirmed Rodriguez’s
worst fear and referred her to Dr. David
D. Wright at FCS. Seeing Wright for
treatment, Rodriguez said, was “the best
decision ever, hands down. I felt very
comfortable with him and with his patient
care staff. The way they welcomed me made
me feel like I was a part of their family.”
Wright, who sees patients at FCS’s
Brandon Cancer Center, New Tampa
and the Tampa Cancer Center, attended
medical school at the University of South
Florida’s Moffitt Cancer Center. It was
during his first year of school that Wright’s
mother received her own cancer diagnosis.
“My mother’s oncologist treated her
like she was family,” Wright told FCS in a
video testimony. “He spent time with her
like she was the only person that mattered
to him, and I’m sure he had about 20
other patients waiting. Her outcome
was very good; he became a friend of the
family and she’s still with us. I realized the
important thing, for me, was the personal
touch. Now, I have patients that have
become part of my life.”
Rodriguez remembers how, at her very
first visit with Wright, he called her by her
first name, as if they were already friends.
Wright took the time to get to know
her, she said, and ensured that she was
comfortable from the very beginning.
“When I walked in for treatment every
day after, I was always greeted with a
smile,” Rodriguez said. “From the front
door staff to the individuals who put in my
IV, everyone was amazing.”
BY HANNAH BURKE
Conquering
Breast Cancer
Warrior spirit helped Amy Rodriguez
stay positive
PATIENT SPOTLIGHT
Spring 2022 15
PATIENT SPOTLIGHT
One day, Rodriguez took it upon herself,
while undergoing chemotherapy, to shave her
head. She continued filling in her eyebrows
and doing her makeup, but she still grappled
with the insecurities that follow such a drastic
change. What if she was now unrecognizable?
Turns out, at Rodriguez’s next appointment,
an FCS staff member would supply her with
a much-needed boost of confidence.
“I can’t recall her name, but I remember
coming in and seeing her look at me with
just the widest smile,” Rodriguez said. “Her
eyes were just gleaming, and she told me,
‘You are so beautiful.’ She remembered me.”
After months of treatment, a double
mastectomy and reconstructive surgery,
Rodriguez said she’s “feeling great — as
though I’m back to myself.” She finished
her last round of treatment in May 2021,
is forging ahead with her military career,
and reports that, despite everything, she
continues to pass her physical fitness tests.
Rodriguez is, to many, a hero.
Last November, she was officially
recognized as such by the Tampa Bay
Buccaneers at Raymond James Stadium.
Prior to the Bucs’ home game against the
Chicago Bears, Rodriguez, the “Hero of
the Game,” received the traditional honor
of “rallying the Krewe” by ringing the
bell of the team’s pirate ship. She beamed,
danced and beckoned the crowd to match
her enthusiasm.
For Rodriguez, that fervor isn’t reserved
just for game days, but for every day she
can continue living, serving others and
loving her family.
“I 100% believe that Florida Cancer
Specialists helped me beat cancer,”
Rodriguez said. “Thank you, care team.
Thank you for loving me through both
your actions and words, and thank you for
making this experience something I will
never forget.”
“I 100% believe
that Florida Cancer
Specialists helped me
beat cancer…Thank you,
care team. Thank you
for loving me through
both your actions and
words, and thank you for
making this experience
something I will
never forget.”
Traveling the world, U.S. Army National Guard Staff
Sergeant Amy Rodriguez has built a lifetime of
unforgettable experiences.
16 FCS Magazine
BY HANNAH BURKE
Data
Governance
Company-wide initiative will provide
standards for collection and storage
ast year, Florida Cancer Specialists enthusiastically
launched its Data Governance Program, an initiative
designed to institute policies, processes and standards
for how data is collected, organized, stored and used
throughout the organization.
The FCS Data Governance Committee was established
to oversee the program’s implementation and monitor its
progress. FCS Chief Executive Officer Nathan H. Walcker
serves as executive sponsor of the committee and Vice
President of Informatics Trevor Heritage, PhD has taken on
the role of committee chair.
It was important to FCS that the committee include
representatives from the organization’s teams and
departments. Data plays a role in every sector of FCS
operations, so this initiative impacts everyone.
For instance, FCS data may also correlate with how
clinical interventions and observations are documented, or
the way in which patient information is attained at intake
and later stored, analyzed and secured. Data can relate to
billing and claims, or even include the acronyms used by
FCS to refer to its clinic locations.
“Like other organizations of our size, complexity and
structure, there are many possible ways in which the
cross-system exchange of data and information could be
inconsistent,” Heritage said. “So, it is vital that we have a
plan in place to strengthen the management of our data and
ensure that it is reliable, secure and being properly used. By
doing this, we can more efficiently and deliberately use our
organization’s data to continue providing world-class cancer
care for our patients.”
Spring 2022 17
DATA GOVERNANCE
For more than three decades, FCS
has been dedicated to its mission of
providing communities across Florida with
premier cancer care and treatment. As the
organization continues to grow and evolve,
FCS believes it is imperative that its data
infrastructure, practices and protocols
follow suit.
Walcker said employees will be kept
apprised of developments in its new “data
driven culture” via updates from the Data
Governance Committee members, who
will serve as the program’s central decision
makers. They plan to furnish reports on
process improvement milestones and
contemporary examples of the program’s
impact across FCS.
In addition to the Data Governance
Committee, subject matter users and experts
who frequently use and interact with data
comprise the FCS Data Stewards Council.
They will work to break down data silos
by identifying the best ways for individual
departments to gather, manage and implement
data, as well as how to effectively receive and
apply data created by other FCS branches.
While the framework for data governance
has been set, it will take time, patience
and cooperation from all team members
to realize the program’s full vision. The
Data Governance Committee has already
identified both short- and long-term goals,
and, Walcker said, “Its progress will be
frequently monitored and communicated.”
“By doing this, we can more efficiently and
deliberately use our organization’s data to continue
providing world-class cancer care for our patients.”
18 FCS Magazine
BY EMMA WITMER
Strategic
Partnership
FCS and COA advance
interests of patients
uilding a better system for cancer
care is a job too big for one
organization, no matter how
dedicated its staff may be.
Given that reality, FCS partners with
organizations such as the Community
Oncology Alliance (COA).
Rose Gerber is Director of Patient
Advocacy and Education for COA’s
Patient Advocacy Network (CPAN)
that works closely with FCS and other
practices around the country to spearhead
policymaking, innovation and reform of
the cancer care system.
“I just love the FCS chapter,” Gerber
said. “They keep me inspired. A lot of what
I do is educating and mentoring others. I
try my best to keep leaders interested, and
when I have leaders like Beth Wittmer,
Dr. Mike Diaz and Dr. Lucio Gordan,
who are high-ranking within the practice
and truly support advocacy, I know we can
accomplish great things.”
FCS launched its CPAN chapter in
2014, under the joint leadership of then
Director of Care Management Don
Champlain, RN, MHA, and current
Director of Care Management Beth
Wittmer, RN, OCN. The two served as co-
chairs of the chapter for three years until,
sadly, Champlain lost his battle with cancer.
Over the last eight years, Wittmer and
Gerber have worked side by side to educate
clinicians, patients, survivors and their
families on the complex system of cancer
care and empower them to act.
As the CPAN chapter leader for FCS,
Wittmer is in constant contact with liaisons
throughout the FCS network to garner
support. She organizes quarterly physician-led
speaking events with patients and families
to provide education and identify what
ADVOCACY SPOTLIGHT
Spring 2022 19
ADVOCACY SPOTLIGHT
legislation is coming up that could be helpful
or detrimental to their care. Even with the
challenges posed by COVID-19 restrictions,
Wittmer has persevered with virtual events.
“Our goal with CPAN is to make
people more aware of what they can do by
standing up and advocating for cancer care
and the difference that it truly makes in the
community setting,” Wittmer said. “We
are always looking for ways to bring more
awareness to the importance of this chapter
and the need for more patients, families
and FCS team members to get involved.
FCS doctors and nurses have added
powerful voices to numerous state and
national CPAN events by identifying
patients willing to share their cancer
stories through the “I AM Community
Oncology” campaign. In addition, FCS
has invited Sen. Rick Scott and Sen. Marco
Rubio to experience FCS facilities through
the “Sit in My Chair” event. Prior to the
pandemic, live visits were hosted in FCS
clinics throughout the state.
Time and again, CPAN advocates from
FCS have espoused the importance of
affordable and easily accessible care and the
value of clinical trials, and they regularly
speak on CPAN patient advisory boards at
statewide events and on Capitol Hill.
One issue that the chapter has worked to
address is the impact of pharmacy benefit
managers on access to medication.
FCS and CPAN have also worked in
tandem to block legislation like the Trump
administration’s Most Favored Nation
proposal, which would have barred at least
20 percent of cancer patients on Medicare
from access to treatment.
“We want to de-complicate these issues
so that patients and their families have a
good understanding of what their rights are
and so that their voices can be heard at the
legislative level,” Wittmer said.
“The work we do with CPAN is so
important,” added FCS President &
Managing Physician Michael Diaz, MD.
“The better we initiate and educate our
patients, the better they can advocate for
themselves. It helps them understand
how and where they can turn for support,
and it helps them understand that they
are not alone in this process. If we can
work together, we can help them navigate
through what can be one of the most
difficult times in anyone’s life.”
“Our goal with CPAN
is to make people more
aware of what they can
do by standing up and
advocating for cancer
care and the difference
that it truly makes in the
community setting.”
Top: Dr. Lucio Gordan and Alachua County representatives participate in Sit in My Chair,
an opportunity for local representatives to gain an understanding of the community
oncology experience from the patient's perspective.
Bottom: Members of the CPAN committee join together at the last in-person COA
Conference in 2019.
Email the FCS CPAN group for more
information or to learn how you can get
involved at CPAN@FLCancer.com.
20 FCS Magazine
BY HANNAH BURKE
FCS Retains
Top Ranking
Among OCM
Participants
n 2016, the Center for Medicare & Medicaid
Innovation (CMMI) developed the Oncology
Care Model (OCM), a national payment
program that provides high-caliber, efficiently
managed cancer care at a lower price to Medicare
beneficiaries.
FCS has been enrolled in the program since its
inception, and according to CMMI’s most recent
OCM reconciliation data, has once again been
ranked at the top among the 126 participating
oncology practices across the country.
With its community-based and personalized
practices, state-of-the-art clinical trial offerings and
dedication to offering patients individualized, world-
class cancer care, FCS’s ranking comes as no surprise
to FCS Chief Executive Officer Nathan H. Walcker.
“Our value-based practice initiatives at FCS
are programmatic, intentional and thoughtfully
designed to be organized squarely around the
patient, yielding consistent results that outpace
industry benchmarks and make good on our
commitment to prioritize patient outcomes and
quality,” Walcker said.
“I could not be prouder of our entire team and
their steadfast commitment to delivering value-based
oncology care in communities across Florida.”
FCS provides care for more than 22,000 OCM
beneficiaries annually and has recorded more than
VALUE-BASED CARE PROGRAM