Breast Cancer Diagnosis, Prognostic Factors and Treatment Considerations for The Role of

For Decision Makers in Managed Care
Quarter 3 2013
Diagnosis, Prognostic Factors and
Treatment Considerations for
Breast Cancer
The Role of Genetic
Testing in Oncology
New Agents Improving
Outcomes for Breast Cancer
Health Disparities
Under the ACA:
A Breast Cancer
Patient Scenario
PRSRT STD
US Postage
PAID
Long Prairie MN
Permit # 871
Takes Flight
For more information
Visit KYPROLIS.com
Onyx, Onyx Pharmaceuticals, Onyx Pharmaceuticals logo, Kyprolis and
Kyprolis logo are all trademarks of Onyx Pharmaceuticals, Inc.
©2013 Onyx Pharmaceuticals, Inc., South San Francisco, CA 1112-CARF-436
January 2013
mco
contents
features
Quarter 3 | 2013 | Issue 3
10
Regulatory & Reimbursement
Disparities Remaining Evident Under the Affordable
Care Act
departments
4
Calendar of Events
Dates and locations of upcoming meetings,
workshops and conferences of interest to managed care
oncology professionals.
5
Correspondence
by Kjel A. Johnson, Pharm.D., publisher, ManagedCare Oncology
6
Facts & Figures
Data and accompanying graphics regarding breast cancer.
25
Pipeline Report
New Therapies for Breast Cancer
by Howard “Skip” Burris, M.D., CMO and executive director,
drug development, Sarah Cannon Research Institute
31
Drug & Administration Compendia
Coding, reimbursement and available therapies in
breast cancer.
39
Clinical Trial Update
by John W. Mucenski, B.S., Pharm.D., director of pharmacy
operations, UPMC Cancer Centers
A review of recent clinical trials in breast cancer, including
the methods, results, conclusions and managed care
implications of each trial reviewed.
46
Resources & References
A comprehensive list of breast cancer sources relevant
to managed care oncology professionals.
by Denise K. Pierce, president, DK Pierce & Associates, Inc.
While the intent of the Affordable Care Act is to broaden access to
care, simply having insurance will not end health care disparities.
For many low-income or underinsured women, lack of access to
care is influenced by such things as their contextual understanding
or health care, impact of geography and required cost share.
Addressing such issues will be an ongoing challenge even after the
ACA is fully implemented.
13
Drug Therapy Reviews
Diagnosing, Staging and Treating Invasive Breast Cancer in
the Context of Managed Care
by Steven M. Sugarman, assistant chair, Department of Medicine,
for Clinical Trials in the Regional Network, Memorial SloanKettering Cancer Center
The therapeutic armamentarium for breast cancer has grown,
resulting in more options for clinicians and improved outcomes for
many patients. Meanwhile, testing for inherited susceptibility has
refined the way we identify at-risk patients and bolstered our ability
to provide tailored preventive interventions for breast cancer,
creating an opportunity to save lives and avoid costly treatment
altogether.
20
Industry Thought Leaders
The Current Role and Future Potential of Genetic Testing
in Oncology
ManagedCare Oncology recently spoke with Nicoleta Voian, M.D.,
M.P.H., assistant professor of oncology and director, Clinical
Genetic Service, Department of Medicine, Roswell Park Cancer
Insitute, about the application of genetic testing in managed care
oncology and specifically in the early detection and treatment of
breast cancer.
advertising index
IFC..............................................................................................Onyx
28–30, IBC........................................Magellan Pharmacy Solutions
BC............................................................................................Amgen
managedcareoncology.com
|3
For Decision Makers in Managed Care
calendar of
events
The list of events that follows provides the
dates and loca­tions of upcoming meetings,
workshops and conferences of interest to
managed care oncology professionals.
October
2-5
Association of Community Cancer Centers
National Oncology Conference
Boston, Massachusetts
13-16
American College of Clinical Pharmacy
Annual Meeting
Albuquerque, New Mexico
16-18
Academy of Managed Care Pharmacy 2013
Educational Conference
San Antonio, Texas
21-22
American Society of Health-System Pharmacists
18th Annual Conference for Leaders
in Health-System Pharmacy
Chicago, Illinois
21-23
Self-Insurance Institute of America, Inc. 33rd Annual
National Educational Conference and Expo
Chicago, Illinois
27-30
American Society for Healthcare Risk Management
2013 Annual Conference and Exhibition
Austin, Texas
28-29
Pharmaceutical Care Management Association
Annual Meeting
Rancho Palos Verdes, California
Published by:
Magellan Pharmacy Solutions
6870 Shadowridge Dr., Suite 111
Orlando, FL 32812
Tel: 866-66i-core
Fax: 866-99i-core
[email protected]
www.icorehealthcare.com
Publishing Staff
Media Managers
Erika Ruiz-Colon
Kayla Killian
Advertising and Sales
For information on advertising in ManagedCare Oncology, contact:
Dean Mather
M.J. Mrvica Associates, Inc.
856-768-9360
[email protected]
© 2013 Magellan Pharmacy Solutions, a Magellan Health Company. ManagedCare Oncology
is published in conjunction with Krames StayWell. All rights reserved. All trademarks are
the property of their respective owners. Printed in the U.S.A.
The content of ManagedCare Oncology — including text, graphics, images and information
obtained from third parties, licensors and other material (“Content”) — is for
informational purposes only. The Content is not intended to be a substitute for
professional medical advice, diagnosis or treatment.
ManagedCare Oncology does not verify any claims or other information appearing in any of
the advertisements contained in the publication and cannot take responsibility for any
losses or other damages incurred by readers in reliance on such content.
ManagedCare Oncology Editorial Advisory Board
Chandra P. Belani, M.D.
Miriam Beckner Professor of Medicine, Penn State College of Medicine;
Deputy Director, Penn State Cancer Institute at Penn State Milton S. Hershey
Medical Center
Howard A. “Skip” Burris III, M.D.
Director of Drug Development, Sarah Cannon Research Institute
Richard Cook, Pharm.D.
Pharmacy Manager, Clinical and Quality Programs, Blue Care Network of Michigan
Jeffrey Crawford, M.D.
Chief of Medical Oncology, Duke University Medical Center
William J. Gradishar, M.D.
Professor of Medicine, Director of Breast Medical Oncology, Northwestern
University, Feinberg School of Medicine
Cliff Hudis, M.D.
Chief, Breast Cancer Medicine Service, Memorial Sloan-Kettering Cancer Center
Deborah Partsch, Pharm.D.
Director, Pharmacy Operations, Highmark Blue Cross Blue Shield
Dan Petrylak, M.D.
Associate Professor of Medicine, Columbia University Medical Center
November
3-6
Employer Healthcare and Benefits Congress
Las Vegas, Nevada
December
7-10
American Society of Hematology Annual Meeting
and Exposition
New Orleans, Louisiana
Don’t Miss an Issue!
Online link makes it easy to manage
your subscription.
If you would like to subscribe to ManagedCare Oncology,
please use the link below. If you already receive this
magazine regularly, we’d like you to confirm your
subscription information so you don’t miss an issue.
Visit www.managedcareoncology.com/subscribe-.aspx
and complete the brief form. Then, if anything changes, it
will be easy to update your information.
Thanks, and we hope you find the magazine useful.
|
4
managedcareoncology Quarter 3 2013
correspondence
Chemotherapy Is Going Global
and So Am I …
Goodbye, Readership
If you develop cancer, thank your lucky
stars that you are in this country. Today,
the U.S. has about 316 million people,
while the world has about 7.1 billion,1
so we are 4 percent of the world’s population. However, the U.S. is responsible for
39 percent of the global chemotherapy
spend. Interestingly, the portion of U.S.
chemotherapy spend is decreasing over
time (as shown in Figure 1) ... but why?
The emerging markets, such as Russia,
China and others are now getting access
to these life-saving medications — but
not how you think.
We have discussed the United Kingdom’s National Institute for Health and
Care Excellence (NICE)2 in the past, but
to summarize, it has an annual budget (yes, a budget!) for health care. If a
therapy has a cost per quality-adjusted
life year (QALY) saved of less than
£20,000 (that’s about US$35,000), it is
deemed covered. If greater than £30,000,
it is deemed not covered. When I asked
an English oncologist what they did with
Provenge, he asked, “What drug?” He
hadn’t even heard of it, since it was never
even submitted for coverage review.
Similarly, in 2004 Germany started the
Institute for Quality and Economic Efficiency in Health Care (Institut für Qualität
und Wirtschaftlichkeit im Gesundheitswesen [IQWiG]),3 sometimes referred to as
Figure 1. U.S. Dominates Global Chemotherapy
Sales, but Emerging Markets Are Getting Traction
2002 Onco Sales Split:
2012 Onco Sales Split:
15%
14%
3%
51%
10%
14%
21%
Japan
10%
39%
Pharmerging
24%
ROW
EU5
USA
ROW = rest of world
Source: IMS Health, MIDAS, Dec 2012
the “German NICE.” In this system, assessments of therapies are conducted by the
“Joint Federal Committee” of physicians
and health insurance funds (Gemeinsamer Bundesausschuss [G-BA]). Australia,
in keeping with such a review process, has
the Australian Pharmaceutical Benefits
Advisory Committee. I think these systems
are pretty reasonable, actually.
However, things are vastly different in
other geographies. For example, in Russia,
China and developing countries, only
generics are covered. Interestingly, the
middle class is burgeoning in such emerging markets, and thus the middle class for
the first time can have access to branded
chemotherapies — by paying for them in
cash. There are no subsidies whatsoever.
You can see by these examples that the
extent of provision of chemotherapies
varies wildly from country to country, but
one way or another, coverage is improving. It is pretty interesting stuff, really.
We founded ManagedCare Oncology
five years ago with the intent of providing
timely therapeutic, cost and management
information regarding cancer care to our
readership, which has grown to nearly
20,000 subscribers today. As of today,
I turn the publishing of this fine journal
to the ready and able hands of my colleagues at ICORE/Magellan Health. I am
off to see how this global cancer care
business really works, and hopefully give
insights and tools to those who need to
understand the value of chemotherapy
and caring for their countries’ citizens.
It has been a fun ride … see you down
the road.
Kjel A. Johnson, Pharm.D.
Publisher
ManagedCare Oncology
References
1. U.S. and World Population Clock. United States Census Bureau website. http://www.census.gov/popclock. Accessed July 23, 2013.
2. National Institute for Health and Care Excellence website. http://www.nice.org.uk. Accessed July 23, 2013.
3. Conrad C. The German IQWiG — It’s Not NICE Benefit Assessment in Germany — New Sense or Nuisance? ISPOR Connections.
2006;12(5). http://www.ispor.org/news/articles/oct06/german_policy.asp. Accessed July 23, 2013.
managedcareoncology.com
|5
facts &
figures
In every issue, Facts & Figures provides snapshots of
information key to managed care oncology professionals.
This installment features data regarding breast cancer.
We hope you find these facts and figures of value as you
review your own health plan data.
Magellan Pharmacy Solutions analyzed paid medical claims for health plan members from October 2011 through September
2012 with a breast cancer diagnosis (primary ICD-9-CM diagnosis codes 174 through 175). The following table illustrates
these claims across Medicare and commercial lines of business (LOBs), with an average of approximately $24.3 million in
allowed drug claims per 1 million member lives for those with a breast cancer diagnosis code. Although claims for many
other solid cancers tend to be more heavily weighted toward the Medicare LOB — consistent with an advanced average age at
the time of diagnosis — allowed dollars for breast cancer reflect an incidence of disease that is more evenly distributed across
younger and older demographics.
Medical Claims for Diagnosis Codes 174 through 175 per 1M Lives — Line of Business (LOB)*
LOB Description
Average Allowed per 1M
Average Units per 1M
Average Claims per 1M
Average Members per 1M
$20,745,589.10
657,917
48,342
12,628
Medicare
$27,859,660.63
1,076,225
75,533
18,649
Grand total
$48,605,249.72
1,734,142
123,875
31,277
Commercial
Using the same data, Magellan Pharmacy Solutions analyzed these claims by site of service (SOS), revealing that the vast
majority of breast cancer drug administration services — in terms of average units, claims and members per 1 million — were
received in the physician’s office. This follows the fact that the physician’s office tends to be the most economical site of care
for all stakeholders. The SOS view by request confirms this assertion, with the hospital inpatient/specialty pharmacy and
hospital outpatient settings carrying significantly higher average costs per claim than the physician’s office.
Medical Claims for Diagnosis Codes 174 through 175 per 1M Lives — Site of Service (SOS)*
SOS
Average Members per 1M
HI/SPP
63
Hospital OP
5,248
Average Allowed per 1M
$154,080.76
$9,873,494.20
$139,875.03
Average Units per 1M
Average Claims per 1M
6,298
219
N/A
14,860
Other
423
5,698
652
Physician
7,296
$11,051,651.36
461,895
34,408
Grand total
13,029
$21,219,101.35
473,891
50,139
SOS*
SOS
Average Units per Claim
HI/SPP
29
$702.14
Hospital OP
N/A
$664.44
Other
9
$214.69
Physician
13
$321.19
Grand total
51
$1,902.47
N/A — data integrity issues for units on hospital claims
|
Average $ per Claim
6
managedcareoncology Quarter 3 2013
Among the leading drugs on these breast cancer claims, the humanized monoclonal antibody targeting human epidermal
growth factor receptor 2 (HER2; trastuzumab) featured the highest average allowed claims per 1 million lives. Another biologic
in the top 10 drugs for breast cancer, bevacizumab, previously had a breast cancer indication that has since been revoked by
FDA. Several supportive care agents appear among the leading drugs on these breast cancer claims, including two agents for
bone-related complications (the monoclonal antibody denosumab and zoledronic acid), a colony-stimulating factor (CSF;
pegfilgrastim) and an antiemetic (palonosetron). A number of traditional chemotherapeutics account for the remaining agents
in the top 10 for breast cancer claims — docetaxel, paclitaxel and protein-bound paclitaxel — in addition to ixabepilone (for
disease resistant to paclitaxel) and fulvestrant (for estrogen receptor-positive disease).
Breast Cancer Drug Spend — Average Allowed Claims per 1M Lives for Diagnosis Codes 174 through 175*
Drug
Average
Allowed per 1M
Average
Units per 1M
Average
Claims per 1M
Average
Members per 1M
Injection, trastuzumab, 10 mg
$5,979,314.57
69,117
1,987
228
Injection, pegfilgrastim, 6 mg
$2,799,419.22
736
759
258
Injection, docetaxel, 1 mg
$1,942,627.83
107,548
901
243
Chemotherapy administration, intravenous infusion technique; up to 1 hour,
single or initial substance/drug
$1,281,045.52
4,814
4,887
733
Injection, fulvestrant, 25 mg
$1,008,582.35
10,637
597
109
Injection, paclitaxel protein-bound particles, 1 mg
$1,000,696.34
88,057
437
54
Injection, bevacizumab, 10 mg
$847,302.06
9,974
119
20
Injection, zoledronic acid (Zometa), 1 mg
$624,012.06
2,297
602
196
Injection, denosumab, 1 mg (For billing prior to 1/1/12 use J3590 or C9272)
$616,945.12
37,353
333
113
Injection, palonosetron hydrochloride, 25 mcg
$428,990.25
17,420
1,790
344
Injection, ixabepilone, 1 mg
$299,024.88
3,748
91
18
Injection, paclitaxel, 30 mg
$287,202.37
5,262
862
174
Injection, eribulin mesylate, 0.1 mg (For billing prior to 1/1/12 use J9999 or
C9280)
$245,012.32
2,434
118
22
Therapeutic, prophylactic or diagnostic injection (specify substance or
drug); each additional sequential intravenous push of a new substance/drug
(List separately in addition to code for primary procedure) (Use 96375 in
conjunction with 96365, 96374, 96409, 96413)
$242,049.09
4,482
2,885
536
Chemotherapy administration, intravenous infusion technique; each
additional sequential infusion (different substance/drug), up to 1 hour
(List separately in addition to code for primary procedure) (Use 96417 in
conjunction with 96413)
$230,891.96
1,372
1,228
322
Injection, fosaprepitant, 1 mg
$211,446.46
89,275
640
182
Cyclophosphamide, 100 mg
$206,460.82
12,810
993
306
Injection, gemcitabine hydrochloride, 200 mg
$202,724.80
2,270
297
53
Intravenous infusion, for therapy, prophylaxis or diagnosis (specify substance
or drug); additional sequential infusion of a new drug/substance, up to 1
hour (List separately in addition to code for primary procedure) (Use 96367
in conjunction with 96365, 96374, 96409, 96413 to identify the infusion of
a new drug/substance provided as a secondary or subsequent service after a
different initial service is administered through the same IV access. Report
96367 only once per sequential infusion of same infusate mix.)
$201,254.76
3,731
2,683
454
Injection, carboplatin, 50 mg
$160,537.81
3,640
352
84
managedcareoncology.com
|7
Breast Cancer Drug Spend — Average Allowed Claims per 1M Lives for Diagnosis Codes 174 through 175 (continued)
Drug
Average
Units per 1M
Average
Claims per 1M
Average
Members per 1M
Office or other outpatient visit for the evaluation and management of an
established patient, which requires at least 2 of these 3 key components:
a detailed history; a detailed examination; medical decision making of
moderate complexity. Counseling and/or coordination of care with other
physicians, other qualified health care professionals or agencies are provided
consistent with the nature of the problem(s) and the patient's and/or family's
needs. Usually, the presenting problem(s) are of moderate to high severity.
Typically 25 minutes are spent face-to-face with the patient and/or family.
$154,848.87
1,499
1,539
512
Chemotherapy administration; intravenous, push technique, each additional
substance/drug (List separately in addition to code for primary procedure)
(Use 96411 in conjunction with 96409 and 96413)
$113,222.39
677
632
189
Leuprolide acetate (for depot suspension), 7.5 mg
$107,473.42
263
142
38
Injection, denosumab, 1 mg (Code deleted effective 12/31/11 — see J0897)
$106,980.18
5,628
51
22
Injection, filgrastim (G-CSF), 480 mcg
$90,262.07
195
165
34
Therapeutic, prophylactic or diagnostic injection (specify substance or drug);
subcutaneous or intramuscular
$79,633.66
1,673
1,681
468
Intravenous infusion, for therapy, prophylaxis or diagnosis (specify substance
or drug); initial, up to 1 hour
$78,604.24
643
665
273
Chemotherapy administration, subcutaneous or intramuscular; hormonal
antineoplastic
$75,624.71
1,052
831
170
Injection, doxorubicin hydrochloride, 10 mg
$74,733.10
5,305
530
164
Chemotherapy administration, intravenous infusion technique; each
additional hour (List separately in addition to code for primary procedure)
(Use 96415 in conjunction with 96413)
$72,140.41
1,090
848
272
Office or other outpatient visit for the evaluation and management of an
established patient, which requires at least 2 of these 3 key components:
an expanded problem-focused history; an expanded problem-focused
examination; medical decision making of low complexity. Counseling and/
or coordination of care with other physicians, other qualified health care
professionals or agencies are provided consistent with the nature of the
problem(s) and the patient's and/or family's needs. Usually, the presenting
problem(s) are of low to moderate severity. Typically, 15 minutes are spent
face-to-face with the patient and/or family.
$65,196.12
894
935
357
Chemotherapy administration; intravenous, push technique, single or initial
substance/drug
$63,784.99
376
378
70
Goserelin acetate implant, per 3.6 mg
$56,641.53
159
105
25
Injection, doxorubicin hydrochloride, all lipid formulations, 10 mg (Not
payable by Medicare effective 7/1/12) — see Q2048 or Q2049 (Code deleted
effective 12/31/12) — see J9002
$56,412.89
99
18
5
Injection, darbepoetin alfa, 1 mcg (non-ESRD use)
$50,067.06
10,577
29
10
$1,157,935.13
178,657
19,028
5,970
$21,219,101.35
685,763
50,139
13,029
Drugs with less than $50,000 allowed
Grand total
|
Average
Allowed per 1M
8
managedcareoncology Quarter 3 2013
In looking at claims by diagnosis code, Magellan Pharmacy Solutions found that unspecified female breast cancer carried the
highest average allowed cost, units, claims and members per 1 million lives. These unspecified claims lead by a significant
margin in every category over the next most commonly coded neoplasm, the upper-outer quadrant of the female breast. As
would be expected, male breast cancer diagnoses trailed all diagnoses of female breast cancer in terms of average units, claims
and members per 1 million lives. Male breast cancer diagnoses also featured a significantly lower average allowed cost per
1 million lives than all forms of female breast cancer except for neoplasms of the axillary tail, which registered the lowest
average allowed cost per 1 million lives overall.
Breast Cancer Diagnosis — Average Allowed Claims per 1M Lives for Diagnosis Codes 174 through 175*
Diagnosis
Diagnosis
Code
Malignant neoplasm of breast (female), unspecified
174.9
Malignant neoplasm of upper-outer quadrant of female breast
Average
Units per 1M
Average
Claims per 1M
Average
Members per 1M
$13,897,100.66
442,927
31,705
8,178
174.4
$2,739,355.02
93,623
7,265
1,908
Malignant neoplasm of other specified sites of female breast
174.8
$2,356,574.40
78,608
5,859
1,579
Malignant neoplasm of lower-outer quadrant of female breast
174.5
$534,862.52
16,204
1,232
299
Malignant neoplasm of upper-inner quadrant of female breast
174.2
$475,674.42
15,579
1,141
335
Malignant neoplasm of central portion of female breast
174.1
$467,274.75
16,701
1,339
322
Malignant neoplasm of lower-inner quadrant of female breast
174.3
$411,071.32
10,559
915
200
Malignant neoplasm of other and unspecified sites of male
breast
175.9
$189,698.57
5,464
350
94
Malignant neoplasm of nipple and areola of female breast
174
$136,702.41
5,585
297
95
Malignant neoplasm of male breast
175
$6,429.01
73
8
2
Malignant neoplasm of axillary tail of female breast
174.6
$4,358.27
438
28
17
685,763
50,139
13,029
Total
Average
Allowed per 1M
$21,219,101.35
*Notes:
1. Population includes plans with commercial and Medicare members
2. Date range: October 2011 to September 2012
3. Based on primary diagnosis of 174, 175
4. Outliers excluded
managedcareoncology.com
|9
10
regulatory &
reimbursement
Disparities
Remaining Evident
Under the Affordable Care Act
By Denise K. Pierce, president, DK Pierce & Associates, Inc.
As we all well know, the intent of
the Affordable Care Act (ACA) is to
increase access to care, especially for
those uninsured for one or more
reasons, including (but not limited to):
• Choice to be uninsured
• Lack of employer health benefit
offerings
• Exceeding Medicaid eligibility and
unable to afford health insurance
So, with the advent of the ACA’s
insurance exchanges and Medicaid
expansion, we should be able to
support access to care for all and
ideally see a resulting impact on
patient outcomes, right? Well, let’s
take an example to look at impact on
cancer patients, specifically targeting
breast cancer, for the purpose of this
article. This example may uncover
| managedcareoncology Quarter 3 2013
food for thought as the ACA rolls out
plan enrollment and payors consider
opportunities for their involvement
with the exchanges.
The Devil’s in the Detail
Generally, breast cancer statistics
demonstrate that, from 2000 to 2009
(in the United States), incidence
of breast cancer decreased by
0.9 percent per year among women,
and deaths from breast cancer
decreased significantly, by 2.1 percent.1
A concerted effort that includes
preventive measures, early diagnosis
and the utilization of novel agents and
enhanced regimens has helped reverse
the curve for what was once considered
a much more deadly cancer for women.
As we unravel the statistics, however,
we can see that the results are not so
positive for black women having a
diagnosis of breast cancer.
Breast cancer is the most commonly
diagnosed cancer among black women.
The Centers for Disease Control (CDC)
reported in 2010 that breast cancer was
the leading cause of cancer death for
black women aged 45-64 years.2 What
was most alarming from the CDC data
was that the breast cancer death rate for
these women was 60 percent higher for
black women than white women (see
Figure 1). This is underscored by the
fact that black women also have a lower
five-year survival rate, at 77 percent,
compared to 90 percent for white
women.
Complicating the opportunity to
change the survival trend is that black
women tend to present with advanced
disease. In fact, the CDC estimates that
45 percent of black women present with
advanced-stage disease as compared to
white women, at 35 percent. Education
on self-examination, and the availability
of mammograms and subsequent
Figure 1. Breast Cancer Deaths per
2
100,000
Breastlives
cancer
deaths per 100,000 lives
60
56.8
50
40
30
35.6
20
10
0
White women
Black women
treatment options, has not affected
patient outcomes for black women as it
has for white women.
Back to the ACA
How does this align with access to
care and whether the ACA will have
the effect of broadening that access for
black women? Well, it undoubtedly
requires outreach and education,
because simply having insurance
may not make the difference. A more
recent 2012 CDC study looking at
breast cancer data from 2005 to
2009 demonstrated that, even after
learning that their mammogram is not
normal, 20 percent of black women
delay seeing a doctor for follow-up for
more than 60 days. That’s compared
to 12 percent of white women.3
Under the ACA, patients have access
to mammograms with no cost share.
That is a great step, but one challenge
is how then to ensure the continuity of
follow-up on abnormal mammograms.
Waiting longer for follow-up care may
allow cancer to spread. For a payor, this
can ultimately lead to higher costs for
that patient’s treatment.
The CDC also reported that only 69
percent of black women start treatment
within 30 days of their diagnosis
of breast cancer, compared with 83
percent of white women. This is
among black women who already have
insurance, as well as those who don’t.
Like other types of cancer disparities,
the reasons that black women have
worse breast cancer survival rates
are numerous. They can include
poverty, lack of access to care, lack of
health insurance, literacy barriers and
unequal treatment by professionals
in the health care system. This was
documented in a March 2002 Institute
of Medicine report.4 These variables
are still likely in play in today’s world,
as reflected by the findings in the 2012
CDC data referenced above.
From a payor’s perspective, it’s not
about limiting access to new drugs
and therapies. Yes, there may be prior
authorizations to ensure alignment
with appropriate patients, or specialty
pharmacy distribution oversight, but if
there is data that supports a clinically
relevant outcome for the patient,
payors will generally cover a drug or
regimen. An example that reflects this
is UnitedHealthcare’s episode of care
pilot for oncology (which includes
a payment methodology for breast
managedcareoncology.com
| 11
12
cancer).5 Pertuzumab was approved
by the Food and Drug Administration
(FDA) in June 2012. Although the
episode of care initiative was in
process, and the cost of treating with
pertuzumab was anticipated to be
approximately $180,000, physicians
and UnitedHealthcare agreed that the
drug’s promising clinical trial results
warranted making the treatment
available in the program with payment
outside the episode.
It is also not about limiting access
to care through different settings.
There is, however, greater guidance for
appropriate care through utilization
of pathways that avoids burdening
the health system, while not limiting
cancer care to only the hospital
outpatient or only the physician office
setting. CareFirst’s presentation at this
year’s American Society of Clinical
Oncology (ASCO) meeting reflected
this. CareFirst reported overall savings
from its ongoing clinical pathways
program for oncology, showing that
CareFirst reduced its overall costs for
treating breast, lung and colon cancers
by 15 percent. These savings were
primarily achieved through a
7 percent decline in emergency room
visits, shorter lengths of stay in the
hospital, increased use of generic
medications and more appropriate
use of chemotherapy.
The ACA’s Impact?
Although, on paper, the intent of
the ACA may be to broaden access
to care, it clearly will not end health
disparities — no matter what
the demographics of the patient.
Regrettably, for many low-income or
underinsured women, this access is not
available to them today and may not
be available even under terms of the
ACA, influenced by such things as a
woman’s contextual understanding of
health care, impact of geography and
required cost share — even under the
ACA’s bronze plan.
As an example, let’s take a
nonsmoking, divorced black woman
with a child who makes $35,000 in
annual income, but has no employer
health benefits. Table 1 outlines the
Table 1. Example Silver Exchange Plan Premium Out-of-Pocket
Household income Federal Poverty Level (FPL)
23%
Unsubsidized premium payments required
$4,935
Maximum percent of annual income outlay for premium
7.53%
Final premium payment required by individual (post-subsidy)
$2,788
| managedcareoncology Quarter 3 2013
impact of patient outlay of monies
related to a silver exchange plan.
This example does not take into
account the subsequent cost share that
the individual would be required to
pay, remembering that the silver plan
covers 70 percent of health costs, while
the individual is responsible for the 30
percent component up to a maximum
out-of-pocket. This is still quite a bit of
money for a single mother to outlay.
If she were to move to a bronze plan,
the premium would decrease to $1,943
per year, but the plan itself only covers
60 percent of health costs, requiring
higher upfront coinsurance and
deductible payments.
This article definitely does not
identify ways to resolve the specific
disparities for black women diagnosed
with breast cancer. It is important to
realize that this is only one of many
disparities evident in our health care
system for many ethnicities. This article
does, hopefully, generate food for
thought about some of the outstanding
issues that will not be resolved simply
through legislation such as the ACA.
How those issues are prioritized
and addressed will be the ongoing
challenge.
References
1. Jemal A, Simard EP, Dorell C, et al. Annual report to the nation
on the status of cancer, 1975-2009, featuring the burden and
trends in HPV-associated cancers and HPV vaccination coverage
levels. J Natl Cancer Inst. 2013;105(3):175-201. doi: 10.1093/
jnci/djs491. Published online Jan. 7, 2013.
2. National Vital Statistics System page. Centers for Disease Control
and Prevention website. http://www.cdc.gov/nchs/nvss.htm.
Updated June 6, 2013. Accessed June 7, 2013.
3. Centers for Disease Control and Prevention. Vital signs: Racial
disparities in breast cancer severity — United States, 2005-2009.
MMWR. 2012;61(45):922-926. Published online Nov. 14, 2012.
4. Smedley BD, Stith AY, Nelson AR, eds. Unequal Treatment:
Addressing Racial and Ethnic Disparities in Healthcare.
Washington, DC: The National Academies Press; 2003.
Available at http://www.nap.edu/openbook.php?record_
id=12875&page=R1. Accessed June 7, 2013.
5. Cantlupe J. Bundled payments come to cancer care.
HealthLeaders Media website. March 11, 2013.
http://www.healthleadersmedia.com/page-3/HEP-289965/
Bundled-Payments-Come-to-Cancer-Care. Accessed June 7, 2013.
therapy
reviews
Diagnosing,
Staging
and Treating
Invasive Breast Cancer
in the Context
of Managed Care
by Steven M. Sugarman, M.D., assistant chair, Department
of Medicine, for Clinical Trials in the Regional Network,
Memorial Sloan-Kettering Cancer Center
Trailing only prostate cancer,
breast cancer holds the
distinction of being the most
common non-skin cancer in
the United States and the most
prevalent non-skin cancer
among American women.1
managedcareoncology.com
| 13
14
An estimated 234,580 cases of invasive
breast cancer will be diagnosed
in 2013, with the overwhelming
majority (an estimated 232,340
cases) being diagnosed in women.1
In addition to leading all non-skin
cancer types among women in terms
of prevalence, breast cancer is also
the second leading cause of cancerrelated death in women.1 As such,
breast cancer is expected to account
for 40,030 deaths among women in
2013.1 Accompanying this remarkable
incidence and mortality are significant
treatment costs, giving managed care
stakeholders legitimate reason to take
notice. As of 2010, breast cancer was
associated with the highest direct costs
of all cancer sites, contributing $16.5
billion in medical expenditures to the
national health care budget.2
Although breast cancer occurs
rarely in men, it is approximately 100
times more likely to affect women.3
Age represents another prominent
risk factor, with approximately twothirds of invasive breast cancers
occurring in women aged ≥ 55 years
versus only one out of every eight
invasive breast cancers being detected
in women aged < 45 years.3 Among
the potentially modifiable risk factors
for the disease are weight gain after
age 18, obesity, physical inactivity,
age at the time of first pregnancy and
alcohol consumption.3 Certain medical
findings, such as high breast tissue
density, high bone mineral density and
biopsy-confirmed hyperplasia, likewise
predict a higher risk for breast cancer.
In terms of medical interventions that
have been linked to an elevated risk
for the disease, the use of hormone
replacement therapy (HRT; i.e.,
combined estrogen and progestin) was
identified as being associated with an
increased risk of breast cancer according
to findings from the Women’s Health
Initiative published in 2002.4 However,
after the publication of these results,
the use of HRT significantly decreased,
likely resulting in the nearly 7 percent
reduction in breast cancer incidence
observed from 2002 to 2003.1
Approximately 5 to 10 percent
of breast cancer incidence is directly
attributable to the inheritance of
mutated genes.3 The discovery of
inherited susceptibility syndromes
and the development of various
assays for specific gene mutations
in breast cancer has had profound
implications in the screening, early
detection and treatment of the disease.
The implications of identifying
these mutations are perhaps best
| managedcareoncology Quarter 3 2013
exemplified by those in the BRCA1
and BRCA2 genes, which represent
the most common cause of hereditary
breast cancer. Both of these genes also
portend an increased risk of ovarian
cancer. The risk of breast cancer may be
as high as 80 percent for members of
some families with BRCA mutations,
and presence of a BRCA mutation
can offer prognostic information and
provide an opportunity for preventive
strategies such as prophylactic
mastectomy and/or oophorectomy in
patients identified to be at high risk.3
BRCA-positive breast cancers tend to
occur in younger women and may
be associated with worse prognostic
factors than cancers in women without
one of these gene mutations.3 In terms
of treatment, the presence of a BRCA1
mutation is associated with breast
cancers that tend not to respond to
hormonal treatments, while cancers
associated with the BRCA2 mutation
tend to be hormone-responsive.5
The identification of these gene
mutations and other molecular markers
allows breast cancer to be categorized
into a myriad of different types and
subtypes, but histology remains the
primary means by which the disease is
differentiated. The most common types
of breast cancer are adenocarcinomas.
Invasive ductal carcinoma accounts
for 75 to 80 percent of cases.3 Invasive
lobular carcinoma is the next most
prevalent type, accounting for 10 to
15 percent of cases.3 Less common
adenocarcinoma subtypes include
mucinous, papillary, medullary and
tubular carcinomas. Very rarely, tumors
of the breast can have the appearance
of nonadenocarcinomas, including
squamous cell carcinomas, sarcomas,
lymphomas and small cell carcinomas.
Together, these make up the remaining
~5 to ~10 percent of cases.3 While a
single type of breast cancer usually
exists exclusively in most patients,
occasionally a single breast tumor can
feature a combination of different
types.3 Regardless of the existence of
one or multiple types of breast cancer
in a particular patient, the specific
type(s) of breast cancer present can
have a pronounced impact on the
detection and diagnosis of the disease.
Diagnosis and Staging
Breast cancer is traditionally detected
via mammography or patient selfpalpation; however, the advent
of genetic testing for inherited
susceptibility has revolutionized this
paradigm. While recommendations
from organizations such as the
American Cancer Society (ACS) have
long promoted regular screening
by various methods among
certain demographics (e.g., annual
mammograms for women aged
≥ 40 years, clinical breast exams at
an interval of ≤ 3 years for women in
their 20s and 30s, etc.), genetic testing
allows for more aggressive screening in
previously unrecognized risk groups.3
For example, the ACS recommends
that individuals of any age identified
as having a BRCA mutation, a firstdegree relative with a BRCA mutation
or other breast cancer susceptibility
syndromes who have not undergone
bilateral mastectomy should undergo
annual breast magnetic resonance
imaging (MRI) screening in addition to
mammography.3 Regardless of how a
tumor is detected, a definitive diagnosis
is typically made with an imaging
study-guided biopsy. Imaging can be
performed via mammography (X-ray),
ultrasound or MRI, and the biopsy can
be performed with the assistance of any
of the radiologic techniques in which
the tumor was visualized. Ultrasound
is the preferred image-guided biopsy
method, followed by stereotactic
(i.e., X-ray-guided) and finally MRI.
Ultrasound guidance for biopsies is the
most common and preferred modality
because it is generally simpler and less
expensive. Stereotactic biopsies are
performed for those tumors that can be
visualized mammographically but not
sonographically. MR-guided biopsies
are reserved for those tumors that can
be visualized only via MRI.
Pathologic analyses are used to
determine the type of cancer present
in a specific patient and characterize
the patient’s disease for prognostic
and treatment selection purposes. In
doing so, the pathologist assigns a
grade of 1 to 3 based on the histologic
appearance of the cells and nuclei
and the overall mitotic activity.
Histologic appearance is based on the
arrangement of cells in relation to one
another, which includes the formation
of tubules and their resemblance to
normal cells. Nuclear grade refers
to the appearance of the nuclei (i.e.,
normal to irregular in appearance).
Mitotic activity refers to the number
of cells in the process of dividing.3
Well differentiated tumors generally
resemble normal cells, with few actively
dividing, while poorly differentiated
tumors contain abnormal-appearing
cells and nuclei and are characterized
by rapid growth.3 The patient’s
prognosis worsens with increasing
tumor grade. Because tumor grading is
based on the pathologist’s assessment
of the tumor, it is somewhat subjective
and less consistent than other
prognostic factors. Other analyses
managedcareoncology.com
| 15
16
are more important in determining
prognosis and treatment planning.
Special studies that detect receptor
proteins on the cell surface are
critical in the analysis of invasive
breast cancers. The most important
of these are estrogen receptor (ER)
status, progesterone receptor (PR)
status and human epidermal growth
factor receptor 2 (HER2) status. ER
and PR status are based on whether a
patient’s tumor cells have receptors for
estrogen and progesterone, respectively.
Approximately two-thirds of cases
feature at least one of the two hormone
receptors, with a higher percentage
observed in postmenopausal women.3
Those cancers that test positive for the
presence of either hormone receptor
are considered to be receptor-positive.
They tend to be less aggressive and
generally respond to hormone-based
therapy.3 Similarly designated as either
positive or negative, HER2 status is
based on the level of the growthpromoting HER2 protein present in
the tumor cells. As such, HER2-positive
cancers are characterized by rapid
growth and aggressive spread. Several
medications that target HER2 play an
important role in the management of
these tumors. Multigene assays such
as Oncotype DX® and MammaPrint®
also provide prognostic information.
The Oncotype DX analysis provides
predictive information regarding the
potential benefit of chemotherapy and
has been widely used in determining
whether chemotherapy should be
administered to patients with earlystage, node-negative tumors.
A crucial piece of information in
determining prognosis and assessing
treatment options is determining
the tumor stage. Staging for breast
cancer — as with virtually all
solid tumors — is based on the
conventions of the American Joint
Committee on Cancer (AJCC)
TNM system.3 In this system, “T”
describes tumor characteristics,
“N” represents the presence and
extent of lymph node involvement,
and “M” indicates the existence
of metastases.3 In staging invasive
breast cancer, each of these categories
that can be adequately assessed is
assigned a number indicating the
level of disease involvement, with
0 representing no involvement
| managedcareoncology Quarter 3 2013
and greater numbers representing
more extensive involvement for that
particular category.3 The cumulative
assessment of the numbers assigned
to these categories is known as stage
grouping — a process that ultimately
determines the stage of a patient’s
breast cancer.3 Stages range from
0 to IV, with stage 0 representing
carcinoma in situ (i.e., carcinoma of
the breast that remains in the duct
and has not invaded the surrounding
tissue). Invasive disease encompasses
stages I through IV. Prognosis is
inversely related to stage. The five-year
survival rates for each stage reflect the
worsening prognosis associated with
progressively higher stages (Table 1).3
Stage IV breast cancer indicates that
the tumor has spread to at least one
distant site (beyond the regional lymph
nodes). Although stage IV disease is
rarely curable, it is considered treatable.
The stage of the tumor in combination
with the results of various laboratory
analyses ultimately drives the treatment
strategy employed by clinicians.
Treatment Considerations
Surgery — either breast-conserving
or mastectomy — is the preeminent
first-line treatment option for breast
cancer and is employed at some point
in virtually all cases of the disease
Table 1. American Joint Committee on Cancer (AJCC) TNM Staging System for
Breast Cancer and Corresponding Five-Year Survival Rates3
Stage
Criteria
5-year
survival
rate (%)
0
• Noninvasive disease (e.g., ductal carcinoma in situ [DCIS], Paget disease of the
nipple); no evidence of a primary tumor; no node involvement; no metastases
93
IA
•Tumor is ≤ 2 cm; no node involvement; no metastases
IB
•Tumor is ≤ 2 cm or not found; micrometastases in 1 to 3 axillary lymph
nodes; no metastases
IIA
•Tumor is ≤ 2 cm or not found; spread to 1 to 3 axillary lymph nodes > 2 mm
across OR found in small amounts in internal mammary lymph nodes on
sentinel lymph node biopsy OR spread to 1 to 3 lymph nodes under the arm 81
and to internal mammary lymph nodes; no metastases
OR
•Tumor is > 2 cm across and < 5 cm; no node involvement; no metastases
IIB
•Tumor is > 2 cm across and < 5 cm; spread to 1 to 3 axillary lymph nodes
and/or found in small amounts in internal mammary lymph nodes on
sentinel lymph node biopsy; no metastases
OR
•Tumor is > 5 cm across but does not grow into the chest wall or skin; no
node involvement; no metastases
74
IIIA
•Tumor is ≥ 5 cm across (or cannot be found); spread to 4 to 9 axillary lymph
nodes, or it has enlarged the internal mammary lymph nodes; no metastases
OR
•Tumor is > 5 cm across but does not grow into the chest wall or skin; spread
to 1 to 9 axillary nodes, or to internal mammary nodes; no metastases
67
IIIB
•Tumor has grown into the chest wall or skin; no node involvement OR
spread to 1 to 3 axillary lymph nodes and/or tiny amounts of cancer are
found in internal mammary lymph nodes on sentinel lymph node biopsy
OR spread to 4 to 9 axillary lymph nodes, or it has enlarged the internal
mammary lymph nodes; no metastases
41*
IIIC
•The tumor is any size or can’t be found; spread to 10 or more axillary lymph
nodes OR spread to the lymph nodes under the clavicle OR spread to the
lymph nodes above the clavicle OR involves axillary lymph nodes and
has enlarged the internal mammary lymph nodes OR spread to 4 or more
axillary lymph nodes, and found in small amounts in internal mammary
lymph nodes on sentinel lymph node biopsy; no metastases
49*
IV
•Tumor can be any size; may or may not have spread to nearby lymph nodes;
metastases in distant organs or to lymph nodes far from the breast; the most
common sites of spread are the bone, liver, brain and lung
15
88
*These figures are correct as printed, in that stage IIIB demonstrates worse survival than stage IIIC.
with the exception of stage IV (i.e.,
metastatic) disease. Chemotherapy
and/or hormonal therapy are
frequently used after surgery (i.e.,
adjuvant therapy) to reduce the risk
of recurrence. Radiation therapy is
generally indicated when a patient
undergoes breast-conserving surgery
(i.e., lumpectomy) and in patients
with a high risk of local recurrence
who undergo mastectomy. Sometimes
chemotherapy is employed before
surgery (i.e., neoadjuvant therapy) in
order to shrink the tumor and increase
the likelihood of successful surgery in
patients with locally advanced disease
(i.e., stage III) or in those patients
who can be converted from requiring
mastectomy to permitting lumpectomy
(i.e., breast conservation).
Hormone receptor analyses
determine whether endocrine therapy
should be employed. The addition
of hormone therapy reduces the risk
of relapse in patients with hormone
receptor-positive tumors. This
therapy is often used for 5 to 10 years
depending on the clinical situation.
The available adjuvant hormonal
agents include tamoxifen and the
aromatase inhibitors (e.g., anastrozole,
letrozole and exemestane). Tamoxifen
works by blocking the estrogen
receptor. Aromatase inhibitors work
by reducing estrogen production in
postmenopausal patients but are not
effective in premenopausal patients.
This latter class of agents has shown
an improvement in disease-free
survival but not overall survival in
postmenopausal patients. Therefore,
tamoxifen is generally the treatment
of choice in premenopausal patients,
and aromatase inhibitor therapy or
sequential therapy with tamoxifen
and an aromatase inhibitor is
employed in the adjuvant treatment of
postmenopausal patients.
Hormone therapy is also useful in
patients with stage IV receptor-positive
breast cancer. Generally, the longer
a patient tolerates a given agent, the
more likely she will do well with a
subsequent anti-estrogen. Fulvestrant,
a medication that degrades the
estrogen receptor on receptor-positive
tumors, is often used after failure of
aromatase inhibitors. Interestingly,
everolimus, a drug that blocks mTOR
(the mammalian target of rapamycin),
when combined with exemstane
(a steroidal aromatase inhibitor),
has been effective in treating patients
whose tumors progressed while on
anastrozole or letrozole (nonsteroidal
aromatase inhibitors). In those
with HER2-positive disease, the
targeted biologic agent trastuzumab
(a monoclonal antibody that
targets HER2) is used in addition to
chemotherapy to reduce the risk of
relapse and improve survival. The two
most common combinations used in
this setting are cyclophosphamide and
doxorubicin, followed by paclitaxel
and trastuzumab or trastuzumab
with docetaxel and carboplatin.5 For
managedcareoncology.com
| 17
18
patients with advanced disease who
have not yet received chemotherapy,
pertuzumab in combination with
trastuzumab and docetaxel has been
shown to extend time to progression
compared with trastuzumab and
docetaxel.5 Other targeted agents for
the treatment of metastatic, HER2amplified disease include lapatinib and
ado-trastuzumab emtansine. Approved
in February 2013, this antibodydrug conjugate is a combination of
trastuzumab and mertansine (a tubulin
inhibitor). In patients with HER2positive, unresectable, locally advanced
or metastatic breast cancer who had
previously failed trastuzumab/taxane
therapy, ado-trastuzumab emtansine
improved overall survival by 5.8
months compared with combination
lapatinib/capecitabine therapy.6
In the subset of patients whose
disease is hormone receptor-negative
and HER2-negative — so called
“triple-negative” patients — traditional
chemotherapeutic agents are generally
indicated for adjuvant treatment as well
as in the treatment of metastatic disease.
While these breast cancers generally
respond well to chemotherapy, as
evidenced by the high complete
response rates when administered as
neoadjuvant therapy, they are also often
associated with a poorer prognosis
than other types.3,5 Eventually, virtually
all patients with metastatic breast
cancer will require chemotherapy
regimens that have demonstrated
efficacy in this setting.5 For metastatic
breast cancer, however, combination
chemotherapy appears to offer no
advantage over sequential single-agent
chemotherapy in terms of improving
survival or overall quality of life.7 This
premise was established in a 2003
study of doxorubicin and paclitaxel,
alone and in combination, for the
treatment of metastatic breast cancer by
Sledge et al. and has not been refuted
since.7 Furthermore, combination
chemotherapy is associated with
increased toxicity, causing the
doses of individual agents to be
compromised. Combination therapy
can, however, increase the likelihood
of tumor shrinkage, which may be
important in certain clinical situations
where a rapid response to therapy
is important. Some combinations
have shown improved response rates
compared with single-agent therapy.
Still, in situations in which a rapid
response to chemotherapy is not
crucial (which is usually the case),
the use of sequential, single-agent
chemotherapy is recommended. No
specific sequence of therapy has been
determined to be superior, and the
sequence of chemotherapy is usually
based on previous therapy, anticipated
| managedcareoncology Quarter 3 2013
side effects from treatment and the
patient’s underlying comorbidities.
The most widely prescribed firstline chemotherapy for metastatic
disease is capecitabine, paclitaxel or
docetaxel. Fortunately, for the patient
with advanced breast cancer, many
therapeutic options are available.5
Because the treatment of breast cancer
remains imperfect, clinical trials should
be considered the best treatment option
for appropriate candidates.
Future Perspectives in
Managed Care
The therapeutic armamentarium for
breast cancer has grown significantly
over the past several years, resulting
in more options for clinicians as well
as improved outcomes for patients
in many cases. With more than 100
investigational agents in phase 2 or 3
clinical trials for breast cancer, the future
looks bright.8 As our knowledge of
the molecular mechanisms of the
disease grows, so does our ability to
target specific steps in these pathways.
Included among these promising agents
are small molecules that target tumor
cell spread and enzymes that are very
active in growing cancers, such as PI3
kinase, AKT kinase and RAF kinase.
Coupled with these developments are
pharmacogenetic assays that can help
determine treatment response and
ensure that novel targeted agents are
not inappropriately used in patients
unlikely to benefit from them. The
introduction and exploitation of
trastuzumab and other HER2-targeted
agents has shown the utility of these
assays, but this is merely the proverbial
tip of the iceberg. Also in the discipline
of genetics, testing for inherited cancer
susceptibility has refined the way we
identify at-risk patients and bolstered
our ability to provide tailored preventive
interventions for breast cancer, creating
an opportunity to save lives and avoid
costly treatment altogether.
While these recent developments —
and similar advances on the horizon —
come at significant expense to payors,
managed care stakeholders are generally
cognizant of their value in cost
avoidance and improved outcomes.
As a result, payors are relatively
liberal in their coverage of newer
targeted biologics, pharmacogenetic
testing and germline genetic testing
for breast cancer, provided that these
interventions are used prudently
and appropriately. Furthermore, as
evidence that they are savvy to the
value of pharmacogenetic testing in
driving appropriate use, most payors
have incorporated specific analyses
(e.g., HER2 testing for trastuzumab)
into their utilization management
initiatives by requiring certain tests
prior to authorizing payment of specific
therapies when available.
Management interventions, such
as Prior Authorization programs and
consensus guideline-driven clinical
pathways, are also being used nationally
in an effort to promote the appropriate
use of costly therapies in the treatment
of breast cancer. So long as these
programs are based on the consensus
opinions of leaders in their respective
fields (e.g., evidence-based guidelines
from organizations such as the National
Comprehensive Cancer Network
[NCCN]), they provide a framework
for determining inappropriate use.
However, payors and clinicians alike
must remain aware of the fact that
oncology is just as much an art as it
is a science, and not every patient’s
disease takes the route set forth in
textbooks and journals. Taking this into
consideration, payors should remain
flexible in their coverage policies,
whether they employ Utilization
Management programs or not, so that
clinicians are not handcuffed and
prevented from making evidence-based,
common-sense decisions. A timely
and responsive appeals process within
the plan infrastructure can facilitate
this. Providers are also accountable for
ensuring that the whole process runs
fluidly; that is, with new data emerging
every day, it is incumbent upon
physicians to know how far that data
can take them and the implications
of results from clinical trials on real,
everyday practice. Ultimately, a spirit
of collaboration between payors and
providers is in the best interests of all
parties involved and, given the growing
array of medical interventions available,
presents the optimal scenario for
improved outcomes in the treatment
of breast cancer.
References
1. American Cancer Society. Cancer Facts & Figures 2013. http://www.cancer.org/research/cancerfactsfigures/cancerfactsfigures/
cancer-facts-figures-2013. Accessed July 24, 2013.
2. The Cost of Cancer. National Cancer Institute at the National Institutes of Health website. http://www.cancer.gov/aboutnci/
servingpeople/cancer-statistics/costofcancer. Accessed July 24, 2013.
3. American Cancer Society. Breast Cancer — Detailed Guide. http://www.cancer.org/cancer/breastcancer/detailedguide. Accessed
July 24, 2013.
4. Rossouw JE, Anderson GL, Prentice RL, et al. Risks and benefits of estrogen plus progestin in healthy postmenopausal women:
principal results from the Women’s Health Initiative randomized controlled trial. JAMA. 2002;288:321-333.
5. National Comprehensive Cancer Network. Clinical Practice Guidelines in Oncology: Breast Cancer. http://www.nccn.org/
professionals/physician_gls/pdf/breast.pdf.
6. Verma S, Miles D, Gianni L, et al. Trastuzumab emtansine for HER2-positive advanced breast cancer. N Engl J Med.
2012;367:1783-1791.
7. Sledge GW, Neuberg D, Bernardo P, et al. Phase III trial of doxorubicin, paclitaxel, and the combination of doxorubicin and
paclitaxel as front-line chemotherapy for metastatic breast cancer: an intergroup trial (E1193). J Clin Oncol. 2003;21:588-592.
8. Magellan Pharmacy Solutions. Medical Pharmacy & Oncology Trend ReportTM. 3rd ed. 2012. http://www.icorehealthcare.com/
media/329731/2012_trend_report.pdf. Accessed July 24, 2013.
managedcareoncology.com
| 19
20
industry thought
leaders
The Current Role
and Future Potential
of Genetic Testing in Oncology
A Discussion with
Nicoleta Voian, M.D., M.P.H.
Assistant professor of oncology and director, Clinical Genetics Service,
Department of Medicine, Roswell Park Cancer Institute
The genesis and evolution of genetic
testing technologies and their application in preventive medicine have
changed the face of oncology. Despite
the fact that only approximately
5 percent of all cancers are strongly
hereditary, inherited cancer susceptibility can be significant to individual
patients and their immediate families.1
While some degree of heredity may
exist in the development of all types
of cancer, breast cancer features a relatively pronounced element of inherited susceptibility for which genetic
testing has been advanced in recent
years. Specifically, inherited mutations
in certain breast cancer susceptibility
genes account for an estimated 5 to 10
percent of all female breast cancers and
anywhere from 4 to 40 percent of all
male breast cancers.1 As such, reputable
cancer centers across the country have
implemented genetic testing programs
and even dedicated entire departments
to the application of gene analysis in
the discipline of oncology. The accuracy
of the testing employed in these centers
can be characterized via the concepts of
analytical validity and clinical validity,
which — when coupled with individual disease-specific scenarios — lend
themselves to determining the clinical
utility of genetic analyses for a particular
patient.2 ManagedCare Oncology recently
sat down with Nicoleta Voian, M.D.,
M.P.H., assistant professor of oncology
and director, Clinical Genetics Service,
Department of Medicine, Roswell Park
Cancer Institute, to gain her insights
on the application of genetic testing in
managed care oncology and specifically
in the early detection and treatment of
breast cancer.
| managedcareoncology Quarter 3 2013
MCO: Can you please describe the various roles of genetic testing in contemporary oncology practice?
Dr. Voian: Clinical cancer genetics is a
critical component of modern oncology
and preventive medicine. There are three
distinct categories of molecular genetic
testing in oncology: germline genetic
testing, somatic genetic testing and pharmacogenetic testing. Germline genetic
testing for cancer susceptibility assesses
patients for an inherited gene mutation
in order to determine the risk for cancer
in an individual. Genetic counseling is
strongly recommended before and after
these tests, which are sent to specialized
laboratories for analysis. The key goal of
germline analyses is to offer appropriate
genetic tests for appropriate patients in
order to prevent and/or detect cancer
early. The somatic genetic tests that
oncologists use in collaboration with
pathologists look for mutations in the
cancer tumor that were acquired in order
to estimate an individual’s response
to therapy as a prognostic measure.
Pharmacogenetic testing, on germline
as well as somatic mutations, enables
oncologists to identify patients at risk for
severe treatment toxicity or poor treatment response.
MCO: How does genetic testing impact
the quality of care and the cost of managing the cancer patient?
Dr. Voian: Identifying individuals who
carry a gene mutation that confers a
significantly increased risk for cancer is
important because preventive and/or
early detection options can then be provided to these individuals, potentially
helping them avoid costly treatment.
Also, in families where there is a known
familial gene mutation, other family
members are at risk for carrying the
same mutation and will benefit from
the preventive/early detection measures.
Conversely, identifying family members
who do not carry that mutation will
eliminate unnecessary costs associated
with enhanced screening.
MCO: How does genetic testing fit into
oncology care today?
Dr. Voian: Approximately 5 to
10 percent of all cancers occur due to
an inherited susceptibility resulting
directly from gene mutations.1 These
mutations, usually inherited from a
parent, confer a high risk of multiple
primary cancers occurring at younger
ages, and we typically see multiple
family members who inherit the
cancer-predisposing mutation. Early
identification of hereditary cancer
syndromes benefits patients through
a growing number of preventive care
options available to patients and blood
relatives. The most common hereditary
cancer syndromes known are in breast,
ovarian and colorectal cancers.
Awareness regarding inherited
susceptibility syndromes has been
increasing over the past few years. Identifying and referring the appropriate
patients diagnosed with breast cancer
soon after their diagnosis for genetic
counseling and testing is very important. If a patient is identified as having
a gene mutation associated with an
increased risk for a second breast cancer
and/or other cancers, it will empower
that individual with the information
needed to make the appropriate treatment decisions, such as the type of
breast and/or ovarian surgery necessary.
MCO: Can you describe the key opportunities and challenges in managing
oncology genetic testing?
Dr. Voian: Genetic testing introduces
the concept of personalized hereditary
cancer risk assessment, which offers
crucial information to help make medical management decisions to reduce
cancer risk. This information is important to the patient and his or her family
members and often reduces the anxiety
and stress associated with hereditary
cancer susceptibility — a negative result
from testing is welcome news when
there is a known mutation in the family.
Conversely, testing may introduce anxiety in individuals identified as carrying
a gene mutation. Also, the sensitivity
of genetic testing is not 100 percent;
therefore, some undetected mutations
may exist, and testing does not detect all
causes of hereditary cancer.
Furthermore, genetic testing for
inherited cancer susceptibility syndromes is complex. There are different
methods to assess for a gene mutation,
including sequencing and looking for
deletions or duplications. We need to
carefully offer the most complete test
available for a specific case. Because
there are multiple cancer syndromes that
increase the risk for cancer — such as
hereditary cancer syndromes associated
with an increased risk for breast cancer,
ovarian cancer, colon cancer, etc. — it is
important to establish which syndrome
is the most robust in the differential.
Sometimes we have several inherited
cancer syndromes in the differential and
we can use a next-generation sequencing
panel that will assess for multiple genes
in one test. In other cases, the most
appropriate person to be tested first in a
managedcareoncology.com
| 21
22
family suggestive for an inherited cancer
predisposition is not available for testing. In these situations, testing of at-risk,
unaffected family members will encounter limitations. There is a need to very
carefully assess the family pedigree on
both sides, as there are cases, although
rare, where individuals can inherit a
gene mutation from each parent. We
also encounter challenges when we need
to recommend further medical management in individuals who tested negative
for the genetic tests offered but still have
an increased risk for cancer based on
their family history or clinical findings.
MCO: Can you describe the key challenges and opportunities in managing
oncology genetic testing specifically in
the area of breast cancer?
Dr. Voian: As mentioned previously,
there are situations when we have
multiple inherited cancer syndromes
associated with an increased risk for
breast cancer in the differential. There
are breast cancer panels that allow for
testing of multiple genes in one sample.
BRCA1 and BRCA2 were not historically included in those panels because
one laboratory had the patent on those
specific genes, but the Supreme Court
ruled on June 13, 2013, that human
genes cannot be patented.3 This recent
development opens up the potential for
more comprehensive and inclusive breast
cancer panels and decreased costs for
BRCA-related analyses. Still, for BRCA1
and BRCA2, there are different tests that
are offered, and the most appropriate
is selected on a case-by-case basis. This
is often based on ethnicity, such as the
Ashkenazi Jewish panel, or if there is a
previously identified gene mutation or
family history of breast cancer. The complete BRCA test includes full sequencing
and testing for large genomic rearrangements in both BRCA1 and BRCA2 genes,
which will likely now be available from
a number of different laboratories after
the Supreme Court’s ruling.
MCO: When is a patient referred for
genetic testing? What is the process that
you undertake to determine if a patient
qualifies for genetic testing?
Dr. Voian: There are two types of
patients that we see in the genetics
clinic: affected patients and unaffected
patients. Affected patients have cancer
or a history of cancer, while unaffected
patients have only a family history of
cancer. For a patient who is affected
with cancer, discussion with the breast
surgeon, medical oncologist and/or
radiation therapist will determine the
impact of genetic testing on a patient’s
medical care.
The National Comprehensive
Cancer Network has established criteria
for genetic testing eligibility for both
groups, and several other professional
organizations, such as the American
Society of Breast Surgeons and the
American Society of Clinical Oncology,
also provide specific guidelines
and recommendations to providers
for identification of individuals at
increased risk for hereditary cancer
syndromes.4 Common characteristics
of these patients include an early
onset of cancer, a rare cancer such as
male breast cancer or ovarian cancer,
a known familial mutation, family
members with multiple primary
cancers, two or more relatives with the
same type of cancer on the same side
of the family, ethnicity, or pathologic
findings for certain types of cancers,
such as triple negative breast cancer
before age 60.4 Ultimately, the most
vital piece of information to help
determine whether genetic testing
is warranted is the potential clinical
utility derived from testing and the
benefits for family members with regard
to their cancer risk.
| managedcareoncology Quarter 3 2013
MCO: What are the steps you have to
consider when you see a patient who
appears to qualify for genetic testing?
Dr. Voian: During genetic counseling,
we perform a thorough evaluation.
The individual’s personal and family
medical histories are reviewed, with a
focus on the cancer history and benign
findings that can be associated with
inherited cancer predisposition. We
review the medical records of the patient
and family members if possible, as well
as the results of previous genetic tests
when applicable. A physical evaluation
may also be needed in some cases.
Based on the findings of this workup,
we establish whether genetic testing
is appropriate and what specific tests
should be used in those patients deemed
appropriate candidates. Before any
genetic test is performed, the informed
consent form needs to be signed
by the patients to confirm that they
acknowledge the test being performed
in addition to the benefits, risks and
limitations of the test. They’re also
informed about state and federal genetic
privacy laws that protect the majority of
individuals from discrimination with
regard to group health insurance and
employment.
MCO: How do insurance regulations
play into the process of genetic testing?
Dr. Voian: Although payors are generally cognizant of the validity of genetic
consultation and testing and relatively
liberal in their related coverage policies,
these services may or may not be covered
by insurance. By and large, the coverage
is approached on an individual case-bycase basis, meaning that many health
plans routinely cover genetic counseling
and testing if the test is deemed appropriate. If the genetic test is not covered,
patients often choose to forgo testing
due to the high out-of-pocket costs.
MCO: Do you ever recommend testing
as a private pay service to a patient if
the test is not covered by insurance? If
family history is not known, would you
recommend testing?
Dr. Voian: The recommendation of
a genetic test is based on its clinical
utility and the impact on cancer risk
assessment for family members, not
on the reimbursement. If the testing is
performed primarily for the medical
management of other family members,
the patient is informed about the outof-pocket expense and given the option
to pay if desired. If the family history is
unknown, we base our risk assessment
on personal history of cancer, such as
the type of cancer and age at diagnosis.
MCO: Do you have to get the
testing approved by the payor before
administering the test?
characteristics may warrant only a
single site test for a familial mutation
or a full genetic panel.
Dr. Voian: Many genetic laboratories
will hold the sample until insurance
authorization is obtained and the
patient agrees with any out-of-pocket
expenses that may accompany the test,
after which the analysis is initiated.
MCO: What are the specific types
of genetic testing for breast cancer?
Ovarian cancer?
MCO: Are the analyses expensive?
Dr. Voian: Genetic testing is often
considered expensive, with costs
ranging anywhere from a few
hundred to a few thousand dollars,
depending on the genes tested and
the method of analysis. A patient’s
Dr. Voian: The most common gene
mutations associated with breast
cancer are BRCA1 and BRCA2, which
are associated with hereditary breastovarian cancer (HBOC) syndrome.
Rarer gene mutations include TP53
associated with Li-Fraumeni syndrome,
PTEN associated with Cowden
syndrome, CDH1 associated with
hereditary diffuse gastric cancer and
STK11 associated with Peutz–Jeghers
syndrome.
For ovarian cancer, the most
commonly associated hereditary
syndrome is HBOC and Lynch
syndrome associated with mutations in
the MLH1, MSH2, MSH6, PMS2 and
EPCAM genes.
MCO: What does the BRCA mutation
entail for both men and women?
Dr. Voian: For breast cancer, the
inherited risk associated with a BRCA1
and BRCA2 mutation in females is
45 to 84 percent, compared with an
approximate 12 percent risk in the
general population.5-7 Women who
have already had breast cancer have
an increased risk for a new primary
contralateral breast cancer, up to a
64 percent risk in their lifetime.6 For
ovarian cancer, the inherited risk is
anywhere from 11 to 62 percent with
the BRCA mutation, compared with
approximately 2 percent in the general
population.5-9 For male breast cancer,
the inherited risk associated with a
BRCA2 mutation is up to 8 percent,
compared with a less than 1 percent
risk in the general population.10,11
managedcareoncology.com
| 23
24
The risk of prostate cancer is less
pronounced with an inherited BRCA2
mutation at 20 percent, compared with
16 percent in the general population.12
In some families, an increased incidence
of melanoma and/or pancreatic cancer
may exist with an inherited BRCA
mutation.13
MCO: Describe the results of genetic
testing. How do the results influence
further care?
Dr. Voian: A positive result confirms
the hereditary cancer susceptibility
syndrome and the predisposition
for cancer associated with a specific
gene. The medical management
recommendations follow the
established guidelines, and the options
include surveillance, chemoprevention
and risk-reducing surgeries. Other
family members can carry the same
mutation; therefore, genetic counseling
with or without genetic testing is
recommended.
In the event of a negative result,
if there is a known mutation in the
family, then the individual does
not have the risks associated with
that mutation and will follow the
screening recommendations for the
general population. For a negative
result with no known mutation in
the family, the result is considered
uninformative. Sometimes additional
genetic testing is indicated in these
cases. If, after all genetic testing,
the individual is still negative for a
detectable mutation, the medical
management recommendations are
based on personal and family history
of cancer and clinical symptoms. A
patient may also have a variant of
uncertain significance, which is when
there is a change in the gene identified,
but it is unknown whether that change
is harmful or benign. In this latter
scenario, the medical management is
again based on personal and family
history of cancer and clinical symptoms.
MCO: Is there research being done to
improve genetic testing for cancer?
MCO: How do you typically present
the genetic testing results to the
patient?
Dr. Voian: Yes, there is ongoing
research to identify gene mutations
in rare genes or new genes that are
associated with an increased risk for
cancer. For example, familial breast
cancer research is being performed by
Dr. Mary-Claire King at the University
of Washington in Seattle, and at the
Cleveland Clinic, Dr. Charis Eng is
looking for mutations in the PTEN
gene and at new genes associated with
Cowden syndrome.
Dr. Voian: We schedule an in-person
follow-up consultation to disclose
and discuss the genetic test results and
the impact of those results on their
medical management, in addition to
consultation with family members
tailored to each case.
MCO: How are genetic tests regulated?
Dr. Voian: All laboratories that do
genetic testing and share results must
be Clinical Laboratory Improvement
Amendments (CLIA)–certified. CLIA
certification provides federal standards
for quality, accuracy and reliability
of tests. The clinical validity for some
genetic tests is required by the U.S. Food
and Drug Administration. New York
requires that genetic tests be performed
in laboratories that hold a state permit
for its residents. The clinical utility is
determined by the health care providers
and health insurance companies.2
MCO: Overall, do you think genetic
testing and counseling is saving lives?
Dr. Voian: Absolutely. Identifying
individuals who carry a gene
mutation that predisposes them for
a significant increased risk for cancer
will offer options to prevent and
detect cancer at an earlier stage.
Risk-reducing surgeries such as
mastectomy for breast cancer,
salpingo-oophorectomy for ovarian
cancer and colectomy for colorectal
cancer are ultimately lifesaving
procedures.
References
1. American Cancer Society. Cancer Facts & Figures 2013. Atlanta: American Cancer Society; 2013.
http://www.cancer.org/research/cancerfactsfigures/cancerfactsfigures/cancer-facts-figures-2013. Accessed July 10, 2013.
2. U.S. National Library of Medicine. Handbook: Help Me Understand Genetics – Genetic Testing. Genetics Home Reference
website. http://ghr.nlm.nih.gov/handbook/testing?show=all. Accessed July 10, 2013.
3. Association for Molecular Pathology, et al. vs. Myriad Genetics, Inc., et al., 569 U.S. ___ (2013).
http://www.supremecourt.gov/opinions/12pdf/12-398_1b7d.pdf. Accessed July 10, 2013.
4. National Comprehensive Cancer Network. Clinical Practice Guidelines in Oncology (NCCN Guidelines®). Genetic/Familial HighRisk Assessment: Breast and Ovarian. http://www.nccn.org/professionals/physician_gls/pdf/genetics_screening.pdf.
5. Antoniou A, Pharoah PD, Narod S, et al. Average risks of breast and ovarian cancer associated with BRCA1 or BRCA2
mutations detected in case series unselected for family history: A combined analysis of 22 studies. Am J Hum Genet.
2003;72(5):1117-1130.
6. Ford D, Easton DF, Bishop DT, Narod SA, Goldgar DE. Risks of cancer in BRCA1-mutation carriers: Breast Cancer Linkage
Consortium. Lancet. 1994;343(8899):692-695.
7. King MC, Marks JH, Mandell JB; New York Breast Cancer Study Group. Breast and ovarian cancer risks due to inherited
mutations in BRCA1 and BRCA2. Science. 2003;302(5645):643-646.
8. Finch A, Beiner M, Lubinski J, et al. Salpingo-oophorectomy and the risk of ovarian, fallopian tube, and peritoneal cancers in
women with a BRCA1 or BRCA2 mutation. JAMA. 2006;296(2):185-192.
9. Risch HA, McLaughlin JR, Cole DE, et al. Prevalence and penetrance of germline BRCA1 and BRCA2 mutations in a
population series of 649 women with ovarian cancer. Am J Hum Genet. 2001;68(3):700-710.
10.Thompson D, Easton D; Breast Cancer Linkage Consortium. Variation in cancer risks, by mutation position,
in BRCA2 mutation carriers. Am J Hum Genet. 2001;68(2):410-419.
11.Tai YC, Domchek S, Parmigiani G, Chen S. Breast cancer risk among male BRCA1 and BRCA2 mutation carriers. J Natl Cancer
Inst. 2007;99(23):1811-1814.
12.Cancer risks in BRCA2 mutation carriers: The Breast Cancer Linkage Consortium. J Natl Cancer Inst. 1999;91(15):1310-1316.
13.Genetics of Breast and Ovarian Cancer (PDQ®). National Cancer Institute at the National Institutes of Health website.
http://www.cancer.gov/cancertopics/pdq/genetics/breast-and-ovarian/HealthProfessional. Accessed July 10, 2013.
| managedcareoncology Quarter 3 2013
pipeline
report
New Therapies for
Breast Cancer
by Howard “Skip” Burris, M.D., CMO and executive director, drug development, Sarah Cannon Research Institute
Outcomes continue to improve for breast cancer
patients with the study and development of new
agents, including the 2012 regulatory approvals
of everolimus (Novartis, Afinitor) and pertuzumab
(Roche/Genentech, Perjeta), and the 2013
approval of ado-trastuzumab emtansine (Roche/
Genentech, Kadcyla).
These therapies represent the advances
being made with new antibodies,
antibody-drug conjugates (ADCs)
and small molecules targeting specific
pathways. Strategies are being deployed
against the various subtypes of breast
cancer, such as hormone receptorpositive, HER2 amplified and triple
negative disease.
The use of agents inhibiting the
PI3K/AKT/TOR pathway has been
widely discussed in the treatment
of breast cancer. The results of the
BOLERO-2 phase 3 trial established the
benefit of everolimus, an mTOR inhibitor, in postmenopausal metastatic
breast cancer patients. In this doubleblind, placebo-controlled study of
724 patients randomized 2:1 to receive
exemestane and everolimus or placebo,
the investigational arm achieved sta-
tistical superiority for the primary end
point of progression free survival (PFS).
Median PFS of 10.6 months versus
4.1 months was noted, yielding a hazard ratio (HR) of 0.36 and a p value of
10 minus 15. Median survival has not
been reported, but a trend favoring the
everolimus arm was noted. In addition,
benefits were noted in both visceral
metastases as well as bone lesions, and
patients receiving everolimus maintained their quality of life for a longer
period of time. All subsets of patients
had superior outcomes with the everolimus plus exemestane treatment, and
of particular interest, those women
with multiple prior therapies had more
improvement than those less heavily
pretreated.
The results of BOLERO-3 were
reported at the American Society of
Clinical Oncology 2013 annual meeting
in Chicago, and once again, the investigational arm of vinorelbine, trastuzumab, everolimus was superior to
vinorelbine, trastuzumab, placebo for
the primary end point of progression
free survival. The absolute difference
was 7.0 versus 5.8 months, with
HR 0.78, p < .01. The patients were
heavily pretreated, including lapatinib
exposure for 27 percent of the population. A total of 569 women were randomized 1:1 in the study, and received
vinorelbine weekly at 25 mg/m2, trastuzumab 2 mg/kg weekly after a 4 mg/kg
loading dose, and either everolimus
5 mg po qd or placebo.
The BOLERO-1 study will answer the
question of utilization of everolimus in
the first-line setting for HER2-positive
breast cancer patients. More than 700
breast cancer patients have been randomized 1:1 to receive weekly paclitaxel
80 mg/m2 weekly x 3 q 4 weeks, trastuzumab 2 mg/kg weekly after a 4 mg/kg
loading dose and either everolimus
10 mg po qd or placebo. The primary
end point is PFS with secondary end
point evaluations of survival and clinical
benefit, including quality-of-life parameters. Results are expected later this year
or in early 2014.
managedcareoncology.com
| 25
26
Updated survival results from the
phase 3 CLEOPATRA (CLinical Evaluation Of Pertuzumab And TRAstuzumab)
study showed that the combination
of pertuzumab (Perjeta), trastuzumab
(Herceptin) and docetaxel significantly
extended the overall survival of patients
with previously untreated HER2-positive
metastatic breast cancer (mBC), compared to trastuzumab, chemotherapy
and placebo. Results showed that the
risk of death was reduced by 34 percent
for people who received pertuzumab,
trastuzumab and chemotherapy, compared to those who received trastuzumab and chemotherapy (HR = 0.66;
p = 0.0008). At the time of the analysis,
median overall survival had not yet been
reached in people receiving the pertuzumab triplet combination, as more
than half of these people continued
to survive. Median overall survival was
more than three years (37.6 months) for
people who received trastuzumab and
chemotherapy.
In June 2012, the U.S. Food and
Drug Administration (FDA) approved
pertuzumab (Perjeta) in combination
with trastuzumab and docetaxel for the
treatment of people with HER2positive mBC who have not received
prior anti-HER2 therapy or chemotherapy for metastatic disease, based on the
results of the CLEOPATRA study.
CLEOPATRA is an international,
phase 3, randomized, double-blind,
placebo-controlled study. The study
evaluated the efficacy and safety
profile of pertuzumab combined with
trastuzumab and docetaxel, compared
to trastuzumab and docetaxel plus
placebo, in 808 people with previously
untreated HER2-positive mBC or with
HER2-positive mBC that had come
back after prior therapy in the adjuvant
or neoadjuvant setting.
Patients receiving the combination
of pertuzumab, trastuzumab and chemotherapy had a statistically significant
38 percent reduction in the risk of their
| managedcareoncology Quarter 3 2013
disease worsening or death (PFS,
HR = 0.62; p < 0.0001) compared to
people who received trastuzumab, chemotherapy and placebo. The median
PFS improved by 6.1 months from
12.4 months for people who received
Herceptin and chemotherapy to
18.5 months for those who received
pertuzumab, trastuzumab and
chemotherapy.
Perjeta is designed specifically to
prevent the HER2 receptor from dimerizing or pairing with other HER receptors (EGFR/HER1, HER3 and HER4),
which play a role in tumor growth and
survival. The mechanisms of action of
pertuzumab and trastuzumab complement each other, as both bind to the
HER2 receptor, but to different places.
The first antibody-drug conjugate approved for the treatment of a
solid tumor occurred earlier this year
with the label for ado-trastuzumab
emtansine (Kadcyla, T-DM1, Roche/
Genentech) for patients with metastatic
HER2-positive breast cancer, including patients previously treated with
trastuzumab. The results of the EMILIA
trial established the benefits of T-DM1
versus the approved combination of
lapatinib and capecitabine. More than
900 patients were randomized 1:1 to
receive T-DM1 3.6 mg/kg intravenously
q 3 weeks or lapatinib 1,250 mg po
qd plus capecitabine 1,000 mg/m2 po
daily x 14 days q 21 days. Median PFS
favored T-DM1 with 9.6 months versus
6.4 months (HR 0.65), with objective
response rates of 44 percent versus
31 percent and two-year survival of
65 percent versus 52 percent. Followup analysis of overall survival shows
a statistically significant difference
favoring the T-DM1 arm at 30 months
versus 25 months for lapatinib and
capecitabine. These results are particularly interesting when one considers that the median survival in the
registration study for lapatinib and
capecitabine was 17 months a few years
ago, demonstrating just how quickly
the outcomes for HER2-positive
patients are improving.
T-DM1, or ado-trastuzumab
emtansine, is composed of the parent
antibody trastuzumab plus a stable
thioether linker and the potent cytotoxic DM1, a maytansinoid, which
inhibits tubulin with potency 20-100
times greater than the available vinca
alkaloids. Toxicities have been minimal, with little to no alopecia, reversible thrombocytopenia and infrequent
liver function changes.
The safety and toxicity profile of
the ADC T-DM1 has been best demonstrated in the randomized phase 2 study
comparing T-DM1 to the standard
regimen of docetaxel 75-100 mg/m2 q
3 weeks plus trastuzumab. Median PFS
favored T-DM1 over the standard combination, 14.2 months versus
9.2 months, and objective response
rates of 64 percent versus 58 percent. The duration of response in the
T-DM1 arm exceeded 9 months, largely
attributable to the lack of cumulative
toxicities, which may limit the benefit.
Myelosuppression, GI toxicity and
alopecia were substantially worse in the
docetaxel plus trastuzumab arm.
The MARIANNE study results will
be widely anticipated, as the trial
compares three arms: a standard taxane
plus trastuzumab regimen, pertuzumab
plus ado-trastuzumab emtansine or
ado-trastuzumab emtansine plus
placebo. More than 1,100 patients
were randomized in this global study
of first-line metastatic HER2-positive
breast cancer patients, with initial
results expected in 2014. These results
will have a tremendous impact on the
next round of adjuvant studies in this
patient population.
The antibody-drug conjugate CDX
011, glembatumumab, is targeting
GPNMB, a biomarker overexpressed
in several malignancies, including
breast cancer and melanoma. Particularly exciting were the results from the
EMERGE trial, which revealed that
in the subset of patients with triple
negative breast cancer overexpressing
GPNMB, more than one third had an
objective response. Additional randomized trials are under way to better
understand both the subset of patients
and the magnitude of benefit to be
seen with this ADC.
Great interest was generated at
last year’s San Antonio Breast Cancer Conference with the results of
the randomized study evaluating the
cyclin-dependent kinase (CDK) inhibitor palbociclib (Pfizer, PD-0332991).
Progression free survival favored the
combination of palbociclib plus letrozole over letrozole alone, 26 months
versus 7.5 months. A phase 3 study has
been initiated and the promising agent
has received “breakthrough therapy”
status from the FDA.
The list of PI3K/AKT/TOR pathway
inhibitors continues to expand, with
trials ranging from phase 1 to phase 3
under way, many in combination with
hormonal therapies such as aromatase
inhibitors or fulvestrant. A survey of
www.clinicaltrials.gov indicates that
MLN 0128 (Millennium); GDC 0941
(Genentech); AZD 2461 (Astra Zeneca);
BEZ 235, BYL 179 and BKM 120
(Novartis); and SAR 2455409/XL147
(Sanofi), among others, are being
developed. Single-agent and combination responses have been documented.
Toxicities have largely centered on
dermatitis and mucositis. A primary
question centers on whether the benefit
is confined to those patients with
molecular aberrations in the pathway.
The future has never been brighter
for patients with breast cancer. More
and more therapies that are devoid
of the traditional toxicities seen with
chemotherapy are in development.
Survival is being measured in years
versus months, with the hope that such
advances will transfer quickly to the
adjuvant setting, where the possibility
for cures exists.
managedcareoncology.com
| 27
28
Magellan Corporate headline
Total Drug
can Solutions∞
be two lines
Any Drug, Any Provider, Any Site of Service
The pharmaceutical market is changing with traditional pharmaceutical
costs declining and a drug pipeline rife with high cost specialty drugs.
It is estimated that by 2019, specialty products will account for 50% of
total drug spend in the United States.1 And, patients get their medications
through a variety of channels with little coordination to ensure safe and
affordable treatments.
But among the changes, your critical needs
remain the same: finding ways to manage the
full range of prescription drug costs—while
ensuring quality care. Now there’s Total Drug
Solutions, an integrated system that does it
all for you.
The Total Drug Solutions program manages
traditional pharmaceuticals, medical drug
spend, and specialty drug spend to help
reduce costs and improve the quality of care.
• Flexible, customized solutions in specialty,
medical, and traditional pharmacy
• Transparency with flexible pricing models
that support real cost savings
• Clinical expertise in complex conditions
such as oncology, psychiatry, HIV,
rheumatoid arthritis, and multiple
sclerosis
Our Total Drug Solutions 360° approach
offers:
• Integrated approaches across all benefits
and sites of service, including retail and
specialty pharmacies, provider office, and
hospital facilities
• Over 40 years of PBM experience, while
having processed 190M pharmacy claims
per year
• State-of-the-art analytics, reporting,
and management to lower drug costs and
measure outcomes
• Knowledge from serving millions of lives in
medical pharmacy management
1. ESI Drug Trend Report 2012
| managedcareoncology Quarter 3 2013
Total Drug Solutions utilizes our deep expertise in pharmacy management to provide a
patient centric, 360° approach to manage all drugs ensuring the right drug at the right
amount, the right benefit at the right site of service.
Medical Pharmacy Solutions
Our customizable, conditionspecific programs enable payors
to combine and influence both
medical and pharmacy systems
and are proven to reduce overall
medical pharmacy spend.
• Utilization management and
reimbursement strategies
support physician buy and bill
and reduce overall program costs
• Backed by our clinical experts
and external review boards
• Documented cost savings and
improved treatment compliance
while often improving quality of
care
Specialty Pharmacy Solutions
Our specialty pharmacy solutions
provide formulary management
and rebate programs along
with complete specialty drug
distribution services.
• Patients receive one-on-one
clinical coaching programs
that are designed to improve
outcomes while reducing costs
• State of the art reporting,
analytics and clinical support to
help you target spend and bend
trend
Pharmacy Benefit Management
• Pricing model customization
from pure transparency through
capitated/risk
With more than 40 years of PBM
experience, we bring payors,
prescribers, patients, and
pharmacies together around
the right drugs, right doses, and
right frequencies to maximize
effectiveness and minimize cost.
• Impeccable implementation
record with CMS-certified PBM
systems
• Competitive national pharmacy
network rates
To learn more about how Total
Drug Solutions offers you true
cost savings and patient-centric
management for any drug, any
place and any benefit, visit
TotalDrugSolutions.com or call
866-664-2673. We’re ready to put
our customizable solutions to work
for you.
• Trend and formulary
management
• A focus on specialty drug
management not just
distribution
• Formulary
optimization
programs, including
supplemental
rebate negotiation,
optimizes
manufacturer
contracts for
preferred, lower cost
drugs
ToTAlDrugSoluTionS.coM
P-20rev4 (7/13) ©2012 Magellan Health Services, Inc.
managedcareoncology.com
| 29
Magellan Pharmacy Solutions
Specialty Pharmacy • Medical Pharmacy • Pharmacy Benefit Management
Driving healthier outcomes
as well as a healthy
return on your investment
by managing the unseen
components of drug spend
Serving customers
in 25 states and D.C.
Let us put our experience to work for you
40+ years
Medicaid pharmacy
experience
~9 million
Medical pharmacy
lives covered
190 million
Rx claim transactions
processed annually
4.1 million
Pharmacy calls
handled
Total Drug SolutionsSM
$1.8 billion
Total rebates
managed
Any pharmacy, any benefit, anywhere—
360° integrated approach
TOTALDRUGSOLUTIONS.cOM
|
30
managedcareoncology
Quarter 3 2013
P-60 (7/13) ©2013
Magellan Health Services, Inc.
Drug & Administration
compendia
Treatment of
Breast Cancer
With each publication, ManagedCare Oncology’s Drug & Administration Compendia highlights a single medication
or a group of medications that could be utilized in the management of one of the featured oncology diseases.
This section addresses such topics as:
• Associated ICD-9-CM codes
• Drugs that have been FDA-approved
• Drugs that are compendia-listed for off-label use based on
clinical studies that suggest beneficial use in some cases
• Ancillary medications used in cancer treatment
• Reimbursement and coding information
HCPCS/CPT® codes and code description
Current code price (AWP-based pricing)
Most recent Medicare allowable (ASP + 6%), if applicable
Possible CPT administration codes that can be utilized
with each drug
Associated ICD-9-CM Codes:
174 Malignant neoplasm of female breast
Includes breast (female)
connective tissue
soft parts
Paget’s disease of:
breast
nipple
Use additional code to identify estrogen receptor status (V86.0, V86.1)
Excludes skin of breast (172.5, 173.5)
174.0
Nipple and areola
174.1
Central portion
174.2
Upper-inner quadrant
174.3
Lower-inner quadrant
174.4
Upper-outer quadrant
174.5
Lower-outer quadrant
174.6
Axillary tail
174.8
Other specified sites of female breast
Ectopic sites
Inner breast
Lower breast
Malignant neoplasm of contiguous or overlapping sites of
breast whose point of origin cannot be determined
Midline of breast
Outer breast
Upper breast
174.9
Breast (female), unspecified
175
Malignant neoplasm of male breast
Use additional code to identify estrogen receptor status (V86.0, V86.1)
Excludes skin of breast (172.5, 173.5)
175.0
175.9
Nipple and areola
Other and unspecified sites of male breast
Ectopic breast tissue, male
managedcareoncology.com
| 31
32
FDA-Approved Medications Currently Available to Treat Breast Cancer
Current Code Price
(AWP-Based Pricing)
Effective 7/1/13
Medicare Allowable
(ASP + 6%) — Effective
7/1/13-9/30/13
Possible CPT
Administration Code(s)
generic (Brand) Name
HCPCS Code — Code Description
ado-trastuzumab emtansine
(Kadcyla)
C9131 — Injection, ado-trastuzumab emtansine,
1 mg (See also J9999)
$33.28
$29.40
96413, 96415
ado-trastuzumab emtansine
(Kadcyla)
J9999* — Not otherwise classified,
antineoplastic drugs
NDC-level pricing
NDC-level pricing
96413, 96415
anastrozole (Arimidex)
J8999* — Prescription drug, oral,
chemotherapeutic, Not Otherwise Specified
NDC-level pricing
NDC-level pricing
N/A
anastrozole (Arimidex)
S0170 — Anastrozole, oral, 1 mg
$13.48
S0170 — not payable by
Medicare
N/A
capecitabine (Xeloda)
J8520 — Capecitabine, oral, 150 mg
$11.50
$9.59
N/A
capecitabine (Xeloda)
J8521 — Capecitabine, oral, 500 mg
$38.34
$31.90
N/A
cyclophosphamide (Cytoxan)
J8530 — Cyclophosphamide, oral, 25 mg
$2.53
$0.97
N/A
cyclophosphamide (Cytoxan)
J9070 — Cyclophosphamide, 100 mg
$53.01
$40.38
96409, 96413, 96415
docetaxel (Taxotere)
J9171 — Injection, docetaxel, 1 mg
$18.26
$4.76
96413
doxorubicin HCl
(Adriamycin)
J9000 — Injection, doxorubicin hydrochloride,
10 mg
$12.00
$3.67
96409
epirubicin (Ellence)
J9178 — Injection, epirubicin hydrochloride, 2
mg
$2.69
$1.34
96409, 96413
eribulin (Havalen)
J9179 — Injection, eribulin mesylate, 0.1 mg
$111.48
$97.92
96409
estradiol (Estrace)
J8499*— Prescription drug, oral,
nonchemotherapeutic, Not Otherwise Specified
NDC-level pricing
NDC-level pricing
N/A
everolimus (Afinitor)
C9399* — Unclassified drugs or biological
(Hospital Outpatient Use ONLY)
NDC-level pricing
NDC-level pricing
N/A
everolimus (Afinitor)
J8999* — Prescription drug, oral,
chemotherapeutic, Not Otherwise Specified
NDC-level pricing
NDC-level pricing
N/A
exemestane (Aromasin)
J8999* — Prescription drug, oral,
chemotherapeutic, Not Otherwise Specified
NDC-level pricing
NDC-level pricing
N/A
exemestane (Aromasin)
S0156 — Exemestane, 25 mg
$13.17
S0156 — not payable by
Medicare
N/A
fluorouracil (Adrucil)
J9190 — Injection, fluorouracil, 500 mg
$3.45
$2.12
96409
fulvestrant (Faslodex)
J9395 — Injection, fulvestrant, 25 mg
$105.33
$89.62
96402
gemcitabine (Gemzar)
J9201 — Injection, gemcitabine hydrochloride,
200 mg
$48.00
$5.67
96413
goserelin acetate (Zoladex, 3.6
mg ONLY)
J9202 — Goserelin acetate, implant, per 3.6 mg
$451.19
$189.54
96372, 96402
ixabepilone (Ixempra)
J9207 — Injection, ixabepilone, 1 mg
$79.85
$67.66
96413, 96415
lapatinib (Tykerb)
J8999* — Prescription drug, oral,
chemotherapeutic, Not Otherwise Specified
NDC-level pricing
NDC-level pricing
N/A
letrozole (Femara)
J8999* — Prescription drug, oral,
chemotherapeutic, Not Otherwise Specified
NDC-level pricing
NDC-level pricing
N/A
megestrol (Megace)
J8999* — Prescription drug, oral,
chemotherapeutic, Not Otherwise Specified
NDC-level pricing
NDC-level pricing
N/A
| managedcareoncology Quarter 3 2013
generic (Brand) Name
HCPCS Code — Code Description
Current Code Price
(AWP-Based Pricing)
Effective 7/1/13
megestrol (Megace)
S0179 — Megestrol acetate, oral, 20 mg
$0.66
S0179 — not payable by
Medicare
N/A
methotrexate (Trexall)
J8610 — Methotrexate, oral, 2.5 mg
$3.56
$0.52
N/A
methotrexate sodium
J9250 — Methotrexate sodium, 5 mg
$0.29
$0.27
96372, 96374, 96401,
96409, 96450
methotrexate sodium
J9260 — Methotrexate sodium, 50 mg
$2.85
$2.76
96372, 96374, 96401,
96409, 96450
paclitaxel protein-bound
particles (Abraxane)
J9264 — Injection, paclitaxel protein-bound
particles, 1 mg
$11.42
$9.54
96413
paclitaxel (Taxol)
J9265 — Injection, paclitaxel, 30 mg
$15.54
$4.11
96413, 96415
pertuzumab (Perjeta)
C9292 — Injection, pertuzumab, 10 mg
$116.45
$102.09
96413
pertuzumab (Perjeta)
J9999* — Not otherwise classified,
antineoplastic drugs
NDC-level pricing
NDC-level pricing
96413
tamoxifen (Nolvadex)
J8999* — Prescription drug, oral,
chemotherapeutic, Not Otherwise Specified
NDC-level pricing
NDC-level pricing
N/A
tamoxifen (Nolvadex)
S0187 — Tamoxifen citrate, oral, 10 mg
$1.89
S0187 — not payable by
Medicare
N/A
thiotepa (Thioplex)
J9340 — Injection, thiotepa, 15 mg
$714.00
$265.00
51720, 96409
toremifene citrate (Fareston)
J8999* — Prescription drug, oral,
chemotherapeutic, Not Otherwise Specified
NDC-level pricing
NDC-level pricing
N/A
trastuzumab (Herceptin)
J9355 — Injection, trastuzumab, 10 mg
$90.29
$78.70
96413, 96415
vinblastine (Velban)
J9360 — Injection, vinblastine sulfate, 1 mg
$3.18
$1.62
96409
Medicare Allowable
(ASP + 6%) — Effective
7/1/13-9/30/13
Possible CPT
Administration Code(s)
*When billing a nonclassified medication using a CMS 1500 claim form, you must include both the HCPCS code (e.g., J8999 for Tamoxifen) in column 24D and the drug name, strength and
NDC (National Drug Code) in box 19 or 24A in order to ensure appropriate reimbursement. Please note: Check with payor regarding correct placement of medication information.
managedcareoncology.com
| 33
34
Compendia-Listed Off-Label-Use Medications Currently Available to Treat
Breast Cancer
Current Code Price
(AWP-Based Pricing)
Effective 7/1/13
Medicare Allowable
(ASP + 6%) — Effective
7/1/13-9/30/13
Possible CPT
Administration
Code(s)
generic (Brand) Name
HCPCS Code — Code Description
bacillus Calmette-Guerin
(Tice BCG, TheraCys)
J9031 — Bacillus Calmette-Guerin (BCG) vaccine
(intravesical), per installation
$174.18
$119.30
51720
bacillus Calmette-Guerin
(Tice BCG, TheraCys)
90586 — Bacillus Calmette-Guerin (BCG) vaccine for
bladder cancer, live, for intravesical use
$174.18
$119.30
51720
bacillus Calmette-Guerin
(BCG vaccine)
90585 — Bacillus Calmette-Guerin (BCG) vaccine for
tuberculosis, live, for percutaneous use
$174.18
$119.30
90471, 90472
bevacizumab (Avastin)
J9035 — Injection, bevacizumab, 10 mg
$74.51
$64.64
96413, 96415
carboplatin (Paraplatin)
J9045 — Injection, carboplatin, 50 mg
$86.00
$2.97
96409, 96413,
96415
cisplatin (Platinol AQ)
J9060 — Injection, cisplatin, powder or solution, per
10 mg
$4.33
$1.84
96409, 96413,
96415
dexamethasone (Decadron)
J1100 — Injection, dexamethasone sodium phosphate,
1 mg
$0.15
$0.12
11900, 11901,
20600, 20605,
20610, 96372,
96374
dexamethasone (Decadron)
J8540 — Dexamethasone, oral, 0.25 mg
$0.10
$0.27
N/A
doxorubicin HCI liposomal
(Doxil)
J9002 — Injection, doxorubicin hydrochloride,
liposomal, 10 mg (Code no longer payable by
Medicare effective 7/1/13)
$581.41
N/A
96413
doxorubicin HCI liposomal
(Doxil)
Q2050 — Injection, doxorubicin hydrochloride,
liposomal, 10 mg, Not Otherwise Specified
$581.41
$545.44
96413
etoposide (Vepesid)
J8560 — Etoposide, oral, 50 mg
$66.85
$55.12
N/A
etoposide (Toposar)
J9181 — Injection, etoposide, 10 mg
$0.91
$0.75
96413, 96415
hydrocortisone (Solu-Cortef)
J1720 — Injection, hydrocortisone sodium succinate,
up to 100 mg
$2.40
$4.92
96365, 96366,
96372, 96374
hydroxyurea (Hydrea)
J8999* — Prescription drug, oral, chemotherapeutic,
Not Otherwise Specified
NDC-level pricing
NDC-level pricing
N/A
hydroxyurea (Hydrea)
S0176 — Hydroxyurea, oral, 500 mg
$1.28
S0176 — not payable by
Medicare
N/A
ifosfamide (Ifex)
J9208 — Injection, ifosfamide, 1 g
$44.09
$30.27
96413, 96415
irinotecan (Camptosar)
J9206 — Injection, irinotecan, 20 mg
$15.00
$2.25
96413, 96415
leucovorin calcium
(Wellcovorin)
J0640 — Injection, leucovorin calcium, per 50 mg
$7.80
$4.76
96372, 96374,
96409
leuprolide (Eligard, Lupron
Depot)
J9217 — Leuprolide acetate (for depot suspension),
7.5 mg
$493.20
$206.01
96402
leuprolide (Lupron)
J9218 - Leuprolide acetate, per 1 mg
$23.57
$10.50
96402
lomustine (CeeNU)
J8999* — Prescription drug, oral, chemotherapeutic,
Not Otherwise Specified
NDC-level pricing
NDC-level pricing
N/A
lomustine (CeeNU)
S0178 — Lomustine, oral, 10 mg
$10.59
S0178 — not payable by
Medicare
N/A
medroxyprogesterone (DepoProvera)
J1050 — Injection, medroxyprogesterone acetate, 1 mg
$0.26
$0.22
96402
melphalan (Alkeran)
J8600 — Melphalan, oral, 2 mg
$10.14
$8.71
N/A
| managedcareoncology Quarter 3 2013
generic (Brand) Name
HCPCS Code — Code Description
Current Code Price
(AWP-Based Pricing)
Effective 7/1/13
Medicare Allowable
(ASP + 6%) — Effective
7/1/13-9/30/13
Possible CPT
Administration
Code(s)
melphalan (Alkeran)
J9245 — Injection, melphalan hydrochloride, 50 mg
$1,971.72
$1,273.10
96409, 96413
methlyprednisolone (DepoMedrol)
J1020 — Injection, methylprednisolone acetate, 20 mg
$3.78
$3.12
11900, 11901,
20600, 20605,
20610, 96372,
96374
methlyprednisolone (DepoMedrol)
J1030 — Injection, methylprednisolone acetate, 40 mg
$5.84
$2.89
11900, 11901,
20600, 20605,
20610, 96372,
96374
methlyprednisolone (DepoMedrol)
J1040 — Injection, methylprednisolone acetate, 80 mg
$9.52
$5.54
11900, 11901,
20600, 20605,
20610, 96372,
96374
methlyprednisolone (SoluMedrol)
J2920 — Injection, methylprednisolone sodium
succinate, up to 40 mg
$2.32
$1.85
11900, 11901,
20600, 20605,
20610, 96372,
96374
methlyprednisolone (SoluMedrol)
J2930 — Injection, methylprednisolone sodium
succinate, up to 125 mg
$3.50
$2.64
11900, 11901,
20600, 20605,
20610, 96372,
96374
methlyprednisolone (Medrol)
J7509 — Methylprednisolone, oral, per 4 mg
$1.43
$0.66
N/A
mitomycin (Mutamycin)
J9280 — Injection, mitomycin, 5 mg
$67.20
$23.99
96409
mitoxantrone (Novantrone)
J9293 — Injection, mitoxantrone hydrochloride, per
5 mg
$68.24
$37.17
96409, 96413
oxaliplatin (Eloxatin)
J9263 — Injection, oxaliplatin, 0.5 mg
$2.04
$0.88
96413, 96415
pemetrexed (Alimta)
J9305 — Injection, pemetrexed, 10 mg
$69.54
$59.48
96409
prednisone (e.g., Deltasone,
Orasone)
J7506 — Prednisone, oral, per 5 mg
$0.08
$0.03
N/A
prednisolone (e.g., Orapred,
Millipred)
J7510 — Prednisolone, oral, per 5 mg
$7.88
$0.04
N/A
topotecan (Hycamtin)
J8705 — Topotecan, oral, 0.25 mg
$104.02
$86.64
N/A
topotecan (Hycamtin)
J9351 — Injection, topotecan, 0.1 mg
$7.66
$2.13
96413
triptorelin (Trelstar Depot,
Trelstar LA)
J3315 — Injection, triptorelin pamoate, 3.75 mg
$975.89
$192.43
96372, 96402
vincristine (Vincasar PFS)
J9370 — Vincristine sulfate, 1 mg
$6.61
$4.42
96409
vinorelbine (Navelbine)
J9390 — Injection, vinorelbine tartrate, per 10 mg
$27.00
$10.23
96409
zoledronic acid (Zometa)
J3487 — Injection, zoledronic acid, 1 mg (Code no
longer payable by Medicare effective 7/1/13)
$239.56
N/A
96365, 96413
zoledronic acid (Reclast)
J3488 — Injection, zoledronic acid, 1 mg (Code no
longer payable by Medicare effective 7/1/13)
$199.85
N/A
96365, 96374
zoledronic acid (Reclast,
Zometa)
Q2051 — Injection, zoledronic acid, 1 mg, Not
Otherwise Specified (Code becomes effective for
Medicare billing 7/1/13)
$239.13
$196.42
96365, 96413,
96374
*When billing a nonclassified medication using a CMS 1500 claim form, you must include both the HCPCS code (e.g., J8999 for Hydrea) in column 24D and the drug name, strength and NDC
(National Drug Code) in box 19 in order to ensure appropriate reimbursement.
managedcareoncology.com
| 35
36
Ancillary Medications Used in Cancer Treatment
generic (Brand) Name
HCPCS Code — Code Description
Current Code Price
(AWP-Based Pricing)
Effective 7/1/13
Medicare Allowable
(ASP + 6%) — Effective
7/1/13-9/30/13
Possible CPT
Administration
Code(s)
aprepitant (Emend)
J8501 — Aprepitant, oral, 5 mg
$8.49
$6.68
N/A
granisetron (Kytril)
J1626 — Injection, granisetron hydrochloride, 100 mcg
$3.84
$0.68
96374
granisetron (Kytril)
Q0166 — Granisetron hydrochloride, 1 mg oral, FDAapproved prescription antiemetic, for use as a complete
therapeutic substitute for an IV antiemetic at time of
chemotherapy treatment, not to exceed a 24-hour dosage
regimen
$59.01
$2.53
N/A
granisetron (Kytril)
S0091 — Granisetron hydrochloride, 1 mg (for circumstances
falling under the Medicare statute, use HCPCS code Q0166)
$59.01
S0091– not payable by
Medicare
N/A
ondansetron (Zofran)
J2405 — Injection, ondansetron hydrochloride, per 1 mg
$0.45
$0.16
96372, 96374
ondansetron (Zofran)
Q0162 — Ondansetron, 1 mg, oral, FDA-approved
prescription antiemetic, for use as a complete therapeutic
substitute for an IV antiemetic at the time of chemotherapy
treatment, not to exceed a 48-hour dosage regimen
$6.05
$0.04
N/A
ondansetron (Zofran)
S0119 — Ondansetron, oral, 4 mg (for circumstances falling
under the Medicare statute, use HCPCS code Q0162)
$24.20
S0119 — not payable by
Medicare
N/A
palonosetron (Aloxi)
J2469 — Injection, palonosetron hydrochloride, 25 mcg
$45.96
$18.93
96374
CPT Administration Code Descriptions
CPT Administration Code
Code Description
51720
Bladder instillation of anticarcinogenic agent (including retention time)
96401
Chemotherapy administration, subcutaneous or intramuscular; nonhormonal antineoplastic
96402
Chemotherapy administration, subcutaneous or intramuscular; hormonal antineoplastic
96409
Chemotherapy administration, intravenous, push technique; single or initial substance/drug
96413
Chemotherapy administration, intravenous infusion technique; up to 1 hour, single or initial substance/drug
96415
Chemotherapy administration, intravenous infusion technique; each additional hour (List separately in addition to code for
primary procedure.) (Use 96415 in conjunction with 96413.)
96422
Chemotherapy administration, intra-arterial; infusion technique, up to 1 hour
96423
Chemotherapy administration, intra-arterial; infusion technique, each additional hour (List separately in addition to code for
primary procedure.) (Use 96423 in conjunction with 96422.)
96425
Chemotherapy administration, intra-arterial; infusion technique, initiation of prolonged infusion (more than 8 hours), requiring
the use of a portable or implantable pump
96450
Chemotherapy administration, into CNS (e.g., intrathecal), requiring and including spinal puncture
96365
Intravenous infusion, for therapy, prophylaxis or diagnosis (specify substance or drug); initial, up to 1 hour
96366
Intravenous infusion, for therapy, prophylaxis, or diagnosis (specify substance or drug); each additional hour (List separately in
addition to code for primary procedure.) (Use 96366 in conjunction with 96365, 96367.)
96369
Subcutaneous infusion for therapy or prophylaxis (specify substance or drug); initial, up to 1 hour, including pump setup and
establishment of subcutaneous infusion site(s)
96370
Subcutaneous infusion for therapy or prophylaxis (specify substance or drug); each additional hour (List separately in addition
to code for primary procedure.) (use 96370 in conjunction with 96369.)
96372
Therapeutic, prophylactic or diagnostic injection (specify substance or drug); subcutaneous or intramuscular
96374
Therapeutic, prophylactic or diagnostic injection (specify substance or drug); intravenous push, single or initial substance/drug
References
• HCPCS Level II Expert 2013.
• Current Procedural Terminology (CPT) 2013.
• American Medical Association. ICD-9-CM for Professionals. Vol. 1 and 2. Chicago: AMA Press; 2013.
• RJ Health Systems International, LLC. The Drug Reimbursement Coding and Pricing Guide. Vol. 11, Number 3, Third Quarter 2013.
• FDA-approved indication (product prescribing information).
• www.ReimbursementCodes.com. Powered by RJ Health Systems International, LLC., Rocky Hill, Conn.
• CMS (Centers for Medicare & Medicaid Services) — Medicare Allowable Third Quarter 2013 — Effective Dates 7/1/13 – 9/30/13.
CPT copyright 2013 American Medical Association. All rights reserved. CPT is a registered trademark of the American Medical Association.
This information was supplied by RJ Health Systems International, LLC, located in Rocky Hill, Conn.
| managedcareoncology Quarter 3 2013
Oncology-Related HCPCS Codes
This reference chart will assist the oncology office (office manager, oncology nurse, physician and ancillary staff) and payor with the appropriate codes to utilize when
billing or reimbursing for medication(s).
generic
(Brand)
Name
HCPCS
Code — Code
Description
azacitidine
(Vidaza)
Current
Code Price
(AWP-Based
Pricing)*
Effective
7/1/13
Medicare
Allowable
(ASP + 6%)
— Effective
7/1/139/30/13
Possible
CPT
Admin
Code(s)
FDA-Approved Uses
Compendia-Listed Off-Label Uses
J9025 —
Injection,
azacitidine,
1 mg
Chronic myeloid leukemia (205.1_)
Low-grade myelodysplastic syndrome
lesions (238.72)
High-grade myelodysplastic syndrome
lesions (238.73)
Myelodysplastic syndrome with 5 q
deletion (238.74)
Myelodysplastic syndrome, unspecified
(238.75)
Malignant neoplasm of specified parts of
peritoneum (158.8)
Malignant neoplasm of peritoneum,
unspecified (158.9)
Malignant neoplasm of pleura (163)
Malignant neoplasm of heart (164.1)
Acute myeloid leukemia (205.0_)
Other thalassemia (282.49)
Sickle-cell disease, unspecified (282.60)
Hb-SS disease without crisis (282.61)
Hb-SS disease with crisis (282.62)
$6.59
$5.77
96401
96409
96413
brentuximab
vedotin
(Adcetris)
J9042 —
Injection,
brentuximab
vedotin, 1 mg
Anaplastic large-cell lymphoma
(200.6_)
Hodgkin’s disease (201._)
N/A
$116.16
$102.74
96413
cabazitaxel
(Jevtana)
J9043 —
Injection,
cabazitaxel,
1 mg
Malignant neoplasm of prostate (185)
N/A
$168.16
$139.73
96413
cetuximab
(Erbitux)
J9055 —
Injection,
cetuximab,
10 mg
Malignant neoplasm of lip (140._)
Malignant neoplasm of tongue (141._)
Malignant neoplasm of major salivary
glands (142._)
Malignant neoplasm of gum (143._)
Malignant neoplasm of floor of mouth
(144._)
Malignant neoplasm of other and
unspecified parts of mouth (145._)
Malignant neoplasm of oropharynx
(146._)
Malignant neoplasm of nasopharynx
(147._)
Malignant neoplasm of hypopharynx
(148._)
Malignant neoplasm of other and
ill-defined sites within the lip, oral
cavity and pharynx (149._)
Malignant neoplasm of colon (153._)
Malignant neoplasm of rectum,
rectosigmoid junction and anus
(154._)
Malignant neoplasm of nasal cavities,
middle ear and accessory sinuses
(160._)
Malignant neoplasm of larynx (161._)
Malignant neoplasm of head, neck
and face (195.0)
Secondary and unspecified malignant
neoplasm of lymph nodes of head,
face and neck (196.0)
Malignant neoplasm of trachea, bronchus and
lung (162._)
$61.14
$52.48
96413,
96415
managedcareoncology.com
| 37
38
generic
(Brand)
Name
HCPCS
Code — Code
Description
dactinomycin
(Cosmegen)
Current
Code Price
(AWP-Based
Pricing)*
Effective
7/1/13
Medicare
Allowable
(ASP + 6%)
— Effective
7/1/139/30/13
Possible
CPT
Admin
Code(s)
FDA-Approved Uses
Compendia-Listed Off-Label Uses
J9120 —
Injection,
dactinomycin,
0.5 mg
Malignant neoplasm of
retroperitoneum (158.0)
Malignant neoplasm of bone and
articular cartilage (170._)
Malignant neoplasm of connective and
other soft tissue (171._)
Malignant neoplasm of placenta (181)
Malignant neoplasm of testis (186._)
Malignant neoplasm of kidney, except
pelvis (189.0)
Neoplasm of uncertain behavior of
placenta (236.1)
Malignant melanoma of skin (172._)
Kaposi’s sarcoma (176._)
Malignant neoplasm of ovary (183._)
Malignant neoplasm of vagina (184._)
Malignant neoplasm of penis and other male
genital organs (187._)
Malignant neoplasm of eye (190._)
Complications of transplanted kidney (996.81)
Complications of transplanted heart (996.83)
$705.60
$618.79
96409
ixabepilone
(Ixempra)
J9207 —
Injection,
ixabepilone,
1 mg
Malignant neoplasm of female breast
(174._)
Malignant neoplasm of male breast
(175._)
Malignant neoplasm of prostate (185)
$79.85
$67.66
96413,
96415
panitumumab
(Vectibix)
J9303 —
Injection,
panitumumab,
10 mg
Malignant neoplasm of colon (153._)
Malignant neoplasm of rectum,
rectosigmoid junction and anus
(154._)
Malignant neoplasm of trachea, bronchus and
lung (162._)
$106.90
$91.10
96413
96415
rituximab
(Rituxan)
J9310 —
Injection,
rituximab, 100
mg
Reticulosarcoma (200.0_)
Lymphosarcoma (200.1_)
Burkitt’s tumor or lymphoma (200.2_)
Marginal zone lymphoma (200.3_)
Mantle cell lymphoma (200.4_)
Primary central nervous system
lymphoma (200.5_)
Large cell lymphoma (200.7_)
Other named variants of
lymphosarcoma and reticulosarcoma
(200.8_)
Nodular lymphoma (202.0_)
Leukemic reticuloendotheliosis
(202.4_)
Other malignant lymphomas (202.8_)
Other and unspecified malignant
neoplasms of lymphoid and
histiocytic tissue (202.9_)
Chronic lymphoid leukemia, without
mention of having achieved
remission (204.1_)
Polyarteritis nodosa (446.0)
Wegener’s granulomatosis (446.4)
Rheumatoid arthritis (714.0)
Felty’s syndrome (714.1)
Other rheumatoid arthritis and other
with visceral or systemic involvement
(714.2)
Hodgkin’s paragranuloma (201.0_)
Hodgkin’s granuloma (201.1_)
Hodgkin’s sarcoma (201.2_)
Hodgkin’s disease, lymphocytic-histiocytic
predominance (201.4_)
Acute lymphoid leukemia (204.0_)
Post-transplant lymphoproliferative disorder
[PTLD] (238.77)
Macroglobulinemia (273.3)
Chronic graft-versus-host disease (279.52)
Acute on chronic graft-versus-host disease
(279.53)
Autoimmune hemolytic anemias (283.0)
Von Willebrand’s disease (286.4)
Acquired coagulation factor deficiency (286.7)
Other and unspecified coagulation defects
(286.9)
Immune thrombocytopenic purpura (287.31)
Thrombotic microangiopathy (446.6)
Pemphigus (694.4)
Systemic lupus erythematosus (710.0)
Enlargement of lymph nodes (785.6)
Malignant ascites (789.51)
$779.83
$678.87
96413
96415
* The code price is based on the HCPCS code description. HCPCS (Healthcare Common Procedure Coding System) codes are a component of CMS (Centers for Medicare & Medicaid Services).
The code price is an AWP-based pricing methodology developed by RJ Health Systems International, LLC, Rocky Hill, Conn.
Oncology-Related J-Code References
• HCPCS Level II Expert 2013.
• Current Procedural Terminology (CPT) 2013.
• American Medical Association. ICD-9-CM for Professionals. Vol. 1 and 2. Chicago: AMA Press; 2013.
• Full prescribing information for each drug listed.
• www.ReimbursementCodes.com. Powered by RJ Health Systems International, LLC, Rocky Hill, Conn.
• CMS (Centers for Medicare & Medicaid Services) — Medicare Allowable Third Quarter — Effective Dates 7/1/13 – 9/30/13.
CPT copyright 2013 American Medical Association. All rights reserved. CPT is a registered trademark of the American Medical Association.
This information was supplied by RJ Health Systems International, LLC, located in Rocky Hill, Conn. Current code prices are effective as of 7/1/13.
| managedcareoncology Quarter 3 2013
clinical
trial update
by John W. Mucenski, B.S., Pharm.D., director
of pharmacy operations, UPMC Cancer Centers
New drugs, such as trastuzumab emtansine (T-DM1)
may prove to be invaluable to patients with HER2
(+) disease, while oral antiangiogenic drugs like
sorafenib may play a role in the treatment of
patients with HER2 (-) metastatic breast cancer.
Title: Trastuzumab emtansine for HER2-positive advanced breast cancer.
Authors: Verma S, Miles D, Gianni L, et al.
Reference: N Engl J Med. 2012;367:17831791.
Purpose: Approximately 20 percent
of patients diagnosed with breast
cancer have amplification of human
epidermal growth factor receptor
2 (HER2). This is associated with
a decrease in survival. Combining
monoclonal antibody therapy directed
against HER2 in combination with
standard chemotherapy has been
effective in this patient population.
Studies have shown the combination of trastuzumab (Herceptin)
with standard chemotherapy in the
first-line setting increases the time to
progression and overall survival in
patients with metastatic disease. For
patients who have progressive disease
and have previously been treated with
trastuzumab, an anthracycline and a
taxane, the combination of lapatinib
(Tykerb) and capecitabine (Xeloda)
has become the standard. Trastuzumab emtansine (T-DM1) combines
the HER2-targeting antitumor properties of trastuzumab with the cytotoxic
activity of the microtubule-inhibitory
agent DM1. The antibody and cyto-
toxic agent are conjugated by means
of a stable linker. This allows for intracellular delivery of the cytotoxic drug
specifically to HER2-overexpressing
cells, improves the therapeutic index
and minimizes the exposure of the
cytotoxic agent to normal tissue. Phase
2 studies have shown clinical activity
of this combination in patients with
HER2 (+) advanced breast cancer. This
phase 3 trial will evaluate the efficacy
and safety of T-DM1, in comparison
to lapatinib and capecitabine, in
patients previously treated with trastuzumab and a taxane.
Methods: This randomized, open-label
trial involved patients with HER2 (+),
unresectable, locally advanced or metastatic breast cancer who had received
previous therapy with trastuzumab
and a taxane. Eligible patients had
documented progression of disease, a
left ventricular ejection fraction of at
least 50 percent and an Eastern Cooperative Oncology Group (ECOG)
performance status of 0 or 1. Patients
were randomly assigned in a 1:1 ratio
to receive either T-DM1 or lapatinib
and capecitabine. Those randomized
to receive oral chemotherapy received
lapatinib 1,250 mg per day and
capecitabine 1,000 mg/m2 every 12
hours (maximum planned daily dose
2,000 mg/m2) on days one through 14
of a 21-day schedule. Patients randomly
assigned to T-DM1 received 3.6 mg/kg
intravenously every 21 days. Dose reductions were made secondary to toxicity
in either treatment arm. Patients were
stratified by number of prior therapies
and disease involvement (visceral vs.
nonvisceral). The primary end points
were progression-free survival (PFS),
overall survival (OS) and safety.
Results: A total of 991 patients were
enrolled, with 496 assigned to lapatinib
plus capecitabine and 495 to T-DM1.
PFS was significantly improved in
the patients treated with T-DM1 (9.6
months vs. 6.4 months; hazard ratio
[HR] 0.64; 95 percent confidence interval [CI], 0.55 to 0.77; p < 0.001). The
clinical benefit was consistent across all
clinically relevant subgroups, with a less
definitive benefit among patients aged
≥ 75 years and those with nonvisceral
or nonmeasurable disease. The median
overall survival at the second interim
analysis crossed the stop boundary for
efficacy (30.9 vs. 25.1 months; HR for
death from any cause 0.68; 95 percent
CI, 0.55 to 0.85; p < 0.001). The objective response rate was higher in those
patients treated with T-DM1 (43.6
vs. 30.8 percent); p < 0.001). Serious
adverse events were reported in 88 (18.0
percent) of the patients treated with
lapatinib plus capecitabine vs. 76 (15.5
percent) of those patients treated with
T-DM1. The incidence of grade 3 or 4
adverse reactions was also higher in the
oral chemotherapy group and consisted
managedcareoncology.com
| 39
40
primarily of diarrhea and palmarplantar erythrodysesthesia. The most
common grade 3 or 4 adverse events in
the T-DM1 group were thrombocytopenia and elevated liver enzymes.
Conclusion: T-DM1 significantly prolonged PFS and OS with less toxicity
than lapatinib plus capecitabine in
patients with HER2 (+) advanced breast
cancer previously treated with trastuzumab and a taxane.
Managed Care Implications: Adotrastuzumab emtansine (Kadcyla) has
been FDA-approved as a single agent for
the treatment of patients with HER2 (+)
metastatic breast cancer who previously
received trastuzumab and a taxane, separately or in combination. An interesting
question will be the role of trastuzumab
in patients who progress on this drug.
Title: Phase II randomized study of trastuzumab emtansine versus trastuzumab
plus docetaxel in patients with human
epidermal growth factor receptor 2-positive metastatic breast cancer.
Authors: Hurvitz SA, Dirix L, Kocsis, J, et al.
Reference: J Clin Oncol. March 20,
2013;31(9):1157-1163.
Purpose: HER2 overexpression occurs
in approximately 20 percent of all
breast cancers and is associated with
increased mortality in early-stage disease, decreased time to relapse and an
increased incidence of metastases when
compared to HER2 (-) breast cancer.
HER2-specific targeted therapies have
improved clinical outcomes in patients
with HER2 (+) disease. Single-agent
trastuzumab (Herceptin) has shown
modest activity, but trastuzumab plus
taxane-based therapy has demonstrated
significantly improved overall survival
(OS) and progression-free survival (PFS)
when compared to chemotherapy alone.
Even with these advances, the majority
of patients with HER2 (+) breast cancer
will develop metastatic disease and
progress. Chemotherapy also is associ-
ated with significant toxicity, including
myelosuppression and other adverse
events that can negatively affect a
patient’s quality of life. Trastuzumab
emtansine (T-DM1; Kadcyla) is an
antibody-drug conjugate developed
for the treatment of HER2 (+) cancer.
T-DM1 selectively delivers a cytotoxic
agent intracellularly to tumor cells,
and the targeting antibody, trastuzumab, is itself approved to treat
metastatic breast cancer. Single-agent
studies with T-DM1 reported objective response rates of 32.7 to 44.0
percent in clinical trials, with a low
rate of adverse events. This study
compares the response and tolerability of T-DM1 alone compared to that
of trastuzumab (H) and docetaxel
(Taxotere) (T), a taxane.
Methods: Patients ≥ 18 years of age
with histologically or cytologically
confirmed HER2 (+), unresectable,
locally advanced or metastatic breast
cancer, without prior chemotherapy
or trastuzumab for metastatic disease,
were eligible. Other inclusion criteria
included measurable disease, an
ECOG performance status of 0 or 1
and adequate end organ function. The
| managedcareoncology Quarter 3 2013
study randomly assigned patients in
a 1:1 ratio to either T-DM1 3.6 mg/kg
intravenously once every three weeks or
trastuzumab 8 mg/kg as an intravenous
(IV) loading dose followed by 6 mg/
kg IV every three weeks and docetaxel
75 or 100 mg/m2 IV every three weeks.
Treatment continued until progressive
disease (PD) or unacceptable toxicity.
The primary end points of the study
were investigator-assessed PFS and
safety. Secondary end points included
overall survival (OS), overall response
rate (ORR) and duration of response.
Results: A total of 137 patients were
randomly assigned to receive either HT
(n = 70, of which 68 received therapy)
or T-DM1 (n = 67). The arms were wellbalanced in regard to previous anthracycline therapy and fairly well-balanced in
regard to prior neoadjuvant or adjuvant
trastuzumab or taxane use. The median
duration of therapy was 8.1 months
(range 1 to 29 months) for trastuzumab,
5.5 months (range 0 to 22 months) for
docetaxel and 10.4 months (range 0 to
29 months) for T-DM1. T-DM1 provided
significant improvement in PFS over HT,
with a HR of 0.59 (95 percent CI, 0.36
to 0.97; p = 0.035). The median PFS
was 9.2 months in the HT arm and 14.2
months in the T-DM1 arm (HR 0.59; 95
percent CI, 0.36 to 0.97). The ORR was
58 percent (95 percent CI, 45.5 to 69.2
percent) with HT and 64.2 percent (95
percent CI, 51.8 to 74.8 percent) with
T-DM1. T-DM1 was also found to cause
fewer ≥ grade 3 adverse events (46.4 vs.
90.9 percent), with grade 4 events occurring in 5.8 and 57.7 percent of patients,
respectively. Serious events (25.8 vs.
20.3 percent) and events leading to
discontinuation of therapy (40.9 vs.
7.2 percent) were also less frequent in
those patients treated with T-DM1. The
most common adverse events of any
grade in the HT group were alopecia,
neutropenia, diarrhea and fatigue. In
the T-DM1 group they were fatigue,
nausea, increase in serum AST and
headache. Preliminary OS results were
similar between the treatment arms,
with a median followup of 23 months.
Conclusion: First-line treatment with
T-DM1 for patients with HER2 (+)
metastatic breast cancer provides a
significant improvement in PFS with a
favorable toxicity profile, versus HT.
Managed Care Implications: T-DM1
may prove to be the drug of choice for
first-line therapy in patients with HER2
(+) metastatic breast cancer. Additional
studies will identify its place in the treatment armamentarium.
Title: A double-blind, randomized,
placebo-controlled, phase 2b study
evaluating sorafenib in combination
with paclitaxel as a first-line therapy in
patients with HER2-negative advanced
breast cancer.
Authors: Gradishar WJ, Kaklamani V,
Sahoo TP, et al.
Reference: Eur J Cancer. 2013;49:312-322.
Purpose: Angiogenesis inhibitors have
become important in cancer treatment.
Angiogenesis is associated with disease
progression, tumor growth, invasiveness, metastasis and recurrence in a
number of malignancies including
breast cancer. While monotherapy
with antiangiogenics has shown
only modest activity in patients with
metastatic breast cancer (MBC), when
combined with standard chemotherapy, clinical benefit has been
demonstrated. The combination of
bevacizumab (Avastin) and paclitaxel
(Taxol) in the first-line setting for
MBC showed an improved tumor
response and progression-free survival
(PFS) when compared to single-agent
paclitaxel. Sorafenib (Nexavar) is
a multikinase inhibitor with both
antiproliferative and antiangiogenic activity in hepatocellular and
advanced renal cell carcinomas. Studies using sorafenib in breast cancer
have looked at its use with established
chemotherapy or endocrine treatment
in human epidermal growth factor
receptor 2 (HER2)-negative patients
with the primary end point being
improvement in PFS. These studies led
to mixed results. This study looks at
the efficacy and safety of sorafenib in
combination with first-line paclitaxel
for HER2 (-) advanced breast cancer.
Methods: Patients aged ≥ 18 years
with histologically or cytologically
confirmed HER2 (-) locally recurrent (inoperable) or metastatic breast
cancer who had not been treated with
chemotherapy for advanced disease
were eligible. Other eligibility criteria
included an ECOG performance status
of 0 or 1, and adequate bone marrow,
renal and hepatic function. Patients were
ran­domized in a 1:1 ratio to paclitaxel
90 mg/m2 weekly intravenously on a
three week on, one week off schedule
in combination with placebo or to the
same dosing schedule of paclitaxel in
combination with sorafenib 400 mg
orally twice a day, which was administered continuously. Clinical and
radiologic assessment occurred every
eight weeks for 24 weeks, then every
12 weeks thereafter. The primary end
point was PFS in the intent-to-treat
(ITT) population. Secondary end
points included overall survival (OS),
overall response rate (ORR), duration
of response (DOR) and safety.
Results: A total of 237 patients were
randomized, 119 to paclitaxel plus
sorafenib and 118 to paclitaxel and
placebo. This was the ITT population. A
total of four patients received no study
treatment in the paclitaxel and sorafenib
arm, leaving 115 patients to be evaluated for safety. There was a median PFS
of 6.9 months noted in those patients
treated with sorafenib vs. 5.6 months
in the placebo arm (HR = 0.788; 95
percent CI, 0.558 to 1.112; p = 0.1715).
There was no significant difference in
managedcareoncology.com
| 41
42
OS between the two treatment arms:
16.8 months for the sorafenib arm vs.
17.4 months for those patients treated
with placebo (HR 1.022; 95 percent
CI, 0.715-1.461; p = 0.904). ORR (67.2
vs. 54.2 percent; p = 0.0343) and DOR
(5.6 vs. 3.7 months; p = 0.0157) favored
the sorafenib-treated population and
were statistically significant. Grade 3
or 4 toxicities (sorafenib vs. placebo)
included hand-foot skin reactions (31
vs. 3 percent), neutropenia (13 vs. 7
percent) and anemia (11 vs. 6 percent).
Two treatment-related deaths occurred
in the sorafenib arm.
Conclusion: The addition of sorafenib
to paclitaxel in patients with MBC
improved disease control but did not
significantly improve PFS to support a
phase 3 trial of similar design. Toxicity
of the combination was manageable
with dose reductions.
Managed Care Implications: The activity
of sorafenib with respect to disease control was encouraging, but the PFS results
were inconclusive and more modest
than those reported with paclitaxel
and bevacizumab in advanced breast
cancer. Targeting specific breast cancer
populations where this combination
may provide more definitive clinical
benefit is warranted.
Title: Docetaxel-cisplatin might be
superior to docetaxel-capecitabine in
first-line treatment of metastatic triplenegative breast cancer.
Authors: Fan Y, Xu BH, Yuan P, et al.
Reference: Ann Oncol. 2013;24:12191225.
Purpose: Triple-negative breast cancer
(TNBC), estrogen receptor, progesterone receptor and HER2 negative,
accounts for up to 20 percent of all
breast cancer cases. It has an aggressive
behavior with early visceral metastases and poor outcomes. Numerous
combinations of therapy have been
used in an attempt to improve the
prognosis for patients with TNBC.
This includes optimizing choice and
scheduling of drugs commonly used
to treat the disease, dose intensification, and the introduction of newer,
novel agents such as anti-EGFR and
anti-angiogenesis agents and poly ADP
ribose polymerase (PARP) inhibitors,
but outcomes have been disappoint-
| managedcareoncology Quarter 3 2013
ing. For now, chemotherapy remains the
best therapeutic option in the adjuvant
or metastatic setting in TNBC. A few
retrospective or small neoadjuvant trials have suggested that TNBC may be
more sensitive to DNA-damaging agents
such as cisplatin (Platinol), while other
studies have shown little benefit. Several
phase 2 or 3 studies are presently underway looking at the use of platinumbased therapy in patients with metastatic
TNBC (mTNBC). This article describes
the first prospective, randomized phase
2 clinical trial comparing different regimens in the treatment of mTNBC.
Methods: This prospective, open-label,
randomized phase 2 trial enrolled
women with locally advanced or
metastatic TNBC who had not received
therapy for advanced disease. Patients
were at least 18 years old and had histologically confirmed TNBC, an ECOG
performance status of ≤ 1 and adequate
end organ function. Patients were
randomized in a 1:1 ratio to receive
either docetaxel (Taxotere) 75 mg/m2
and cisplatin 75 mg/m2, both intravenously on day one (TP), or docetaxel
75 mg/m2 intravenously on day one and
capecitabine (Xeloda) orally at a dose
of 1,000 mg/m2 twice a day on a two
weeks on, one week off schedule (TX).
Regimens were repeated every three
weeks for up to six cycles or until disease
progression, unacceptable toxicity or
withdrawal of patient consent. The primary end point was objective response
rate (ORR). Secondary end points
included progression-free survival (PFS),
overall survival (OS) and safety.
Results: A total of 53 patients were
randomized and evaluated, 27 to the
TP arm and 26 to the TX arm. All had
received previous anthracycline therapy,
while over 50 percent in each arm had
received previous paclitaxel (Taxol).
Sixteen of 27 patients (59 percent) in
the TP arm and 19 of 26 (73 percent) in
the TX arm had visceral metastases. The
ORR was 63.0 percent, three complete
responses and 14 partial responses,
in the TP arm and 15.4 percent, four
partial responses in the TX arm (p =
0.001). The median PFS was 10.9
months in those patients treated with
TP (95 percent CI, 2.2-19.8 months)
and 4.8 months in those treated with
TX (95 percent CI, 3.0-6.7 months). The
hazard ratio was 0.29 with a 95 percent
CI of 0.14-0.57 and a p value of < 0.001.
Median overall survival also favored
the patients treated with TP (32.8 vs.
21.5 months, HR 0.41; 95 percent CI,
0.18-0.92; p = 0.027). Overall, both
treatment regimens were well-tolerated.
Grade 3 or 4 neutropenia was similar in
both arms and no grade 3 or 4 thrombocytopenia or anemia was noted. As
expected, gastrointestinal toxicity such
as vomiting (74.1 vs. 34.6 percent;
p = 0.004) was more common in the
patients receiving TP. Likewise, a higher
incidence of grade 2 or 3 hand-foot
syndrome was noted in patients treated
in the capecitabine arm (p = 0.023).
Conclusion: This study suggests that a
cisplatin-based chemotherapy was superior to a capecitabine-based regimen in
the first-line treatment of mTNBC, as
measured by ORR, PFS and OS. Larger
studies need to be completed to evaluate these findings.
Managed Care Implications: With the
lack of efficacy of targeted agents (e.g.,
PARP inhibitors) to effectively treat
patients with mTNBC, other treatment
strategies must be developed. DNA-damaging agents such as cisplatin may be the
cornerstone for further investigations.
Title: Primary results of BEATRICE, a randomized phase III trial evaluating adjuvant bevacizumab-containing therapy in
triple-negative breast cancer.
Authors: Cameron D, Brown J, Dent R,
et al.
Reference: Cancer Res. 2012;72(24
Suppl):Abstract nr S6-5.
Purpose: There are no established
targeted treatment options for patients
with triple-negative breast cancer
(TNBC), a clinically important subset
of breast cancer patients. Studies in
metastatic breast cancer have shown
the addition of bevacizumab (Avastin), an antivascular endothelial
growth factor (anti-VEGF) antibody, to
standard chemotherapy significantly
improves progression-free survival.
Since high VEGF levels have been
observed in estrogen receptor (-) and
progesterone receptor (-) tumors,
bevacizumab may be beneficial in
TNBC based upon its ability to target
the angiogenic switch before tumor
vascularization and the dependency
of micrometastases on angiogenesis.
The objective of this study is to evaluate the addition of bevacizumab to
chemotherapy in the adjuvant setting
for patients with TNBC.
Methods: Patients with centrally confirmed resected, early invasive TNBC
were eligible for the trial. They were
randomized in a 1:1 ratio to receive
either standard chemotherapy (investigator’s choice between taxane-based
therapy, anthracycline-based therapy
or a combination of the two agents).
Bevacizumab was added to half of
the patients at a dose of 5 mg/kg/
week equivalent. Chemotherapy was
administered for four to eight cycles
and bevacizumab was administered
for one year. The primary end point
of the study was three-year invasive
disease-free survival (IDFS). Secondary
end points included interim overall
survival and toxicity.
Results: A total of 2,591 patients were
enrolled, of whom 1,290 received
chemotherapy and 1,301 received
chemotherapy plus bevacizumab.
The three-year IDFS was 82.7 percent in those patients treated with
chemotherapy alone and 83.7 percent
when treated with chemotherapy and
bevacizumab. This was not statistically
significant, with hazard ratio of 0.87
and a p value of 0.181. The interim overall survival data also did not show a statistically significant difference between
the two arms in regard to events (8.3
percent in the chemotherapy alone arm
and 7.1 percent when bevacizumab was
added to chemotherapy [HR 0.84 and p
value of 0.2318]). As expected, grade ≥ 3
adverse events were reported at a higher
frequency in those patients treated with
bevacizumab and chemotherapy (11 vs.
3 percent). Hypertension and venous
thrombotic events were more prevalent
during the treatment phases in both
arms. When bevacizumab was continued by itself following the four to eight
cycles of chemotherapy for the scheduled one year, grade ≥ 3 adverse events
were again higher in those patients
receiving active treatment. The most
common adverse events observed were
congestive heart failure/left ventricular
dysfunction (2 percent), hypertension
(5 percent) and proteinuria (2 percent).
Conclusion: The results of BEATRICE
demonstrated better than anticipated
three-year IDFS in this patient population with early-stage TNBC. There was
no statistically significant improvement
in IDFS with the addition of one year of
bevacizumab to adjunct chemotherapy
for TNBC. Overall, adverse events were
consistent with the established safety
profile of bevacizumab in metastatic
breast cancer.
Managed Care Implications: At first
review of this study, bevacizumab does
not have a role as adjuvant therapy
for patients with triple-negative breast
cancer. However, further followup is
required to assess any potential benefit
of bevacizumab on overall survival.
Title: Efficacy of goserelin plus anastrozole in premenopausal women with
advanced or recurrent breast cancer
refractory to an LH-RH analogue with
managedcareoncology.com
| 43
44
tamoxifen: results of the JMTO BC08-01
phase II trial.
Authors: Nishimura R, Anan K, Yamamoto Y, et al.
Reference: Oncol Rep. May
2013;29(5):1707-1713. doi: 10.3892 or
2013.2312.
Purpose: The majority of all cases of breast
cancer are hormone-receptor (+). Endocrine therapy is used for adjuvant treatment and management of recurrence
in hormone-sensitive breast cancer.
Ovarian suppression induced surgically
or with a luteinizing hormone-releasing
hormone (LH-RH) analog as a postoperative adjunct therapy can prevent
recurrence and prolong survival in this
patient population. In premenopausal
women, estrogen is primarily synthesized by the ovaries. After menopause,
estrogen levels decline and estrogen is
largely produced in peripheral adipose
tissue and in cancer cells. Levels remain
high enough to stimulate the proliferation of breast cancer cells. Therefore, aromatase inhibitors are used as
standard treatment in postmenopausal
women with breast cancer following the
cessation of ovarian function. Endocrine therapy involves the sequential
administration of single agents, but the
combination of an LH-RH analog and
tamoxifen (Nolvadex) is superior to
monotherapy and thus the treatment
of choice for premenopausal women
with advanced or recurrent breast
cancer. Should one become resistant
to this combination, alternatives are
few. Some have recommended that
premenopausal women with advanced
or recurrent breast cancer undergo
ovarian ablation or suppression and
then receive treatment similar to that
recommended for postmenopausal
women, such as an LH-RH analog and
an aromatase inhibitor. However, few
studies support this treatment regime
for premenopausal women. A small
study used goserelin (Zoladex) plus
anastrozole (Arimidex) as second-line
endocrine therapy in premenopausal
women with advanced breast cancer
who had previously received an
LH-RH analog and tamoxifen. After
six months of therapy, the clinical
benefit rate was 75 percent, with the
majority of the patients having stable
disease or a biochemical response.
This study evaluated the response rate
to an LH-RH analog plus anastrozole
in women who failed to respond to an
LH-RH analog and tamoxifen.
Methods: This open-label, single-arm,
multicenter, phase 2 study was open
to premenopausal women aged 20 to
55 with a confirmed diagnosis of metastatic or recurrent estrogen receptor
(+) (ER+) and/or progesterone receptor (+) (PR+) breast cancer. Patients
had to have a measurable lesion or
bony lesion, be refractory to previous
treatment with an LH-RH analog plus
tamoxifen, and have an ECOG performance status of 0 or 1. Treatment was
initiated with anastrozole 1 mg orally
once a day and goserelin, a 3.6 mg
depot injection administered subcutaneously into the lower abdomen every
28 days. Treatment was continued
until the development of progressive disease or unacceptable adverse
events. The primary end point was
objective response rate (ORR), which
was confirmed independently by two
radiologists. Secondary end points
included progression-free survival
(PFS), overall survival (OS), clinical
benefit rate (CBR) and safety.
Results: A total of 37 patients were
enrolled in the trial. The median
age was 43 years (range 33 to 53).
The ER/PR status was ER+/PR+ in
27 patients (73 percent), ER+/PR- in
eight (21.6 percent) and ER-/PR+ in
two (5.4 percent). HER2 was negative
in 36 patients (97.6 percent). During
previous treatment with an LH-RH
analog and tamoxifen, 26 patients
| managedcareoncology Quarter 3 2013
(70.3 percent) had progressive disease,
six (16.2 percent) had recurrence during
postoperative adjuvant therapy and five
(13.5 percent) had completed the previous course of adjuvant therapy. Thirtyone patients had distant metastases and
six had locally advanced disease. One
patient (2.7 percent) had a complete
response (CR) and an additional six
(16.2 percent) had a partial response for
an ORR of 18.9 percent. Sixteen patients
(43.2 percent) had a prolonged stable
disease for a CBR of 62.2 percent (95
percent CI, 44.8-77.5 percent). Eleven
patients (29.7 percent) had progressive disease. The median PFS was 7.3
months with an OS of 35.2 months.
The majority of adverse events were
grade 1 and consisted of hot flashes,
sweating and joint pain.
Conclusion: Goserelin plus anastrozole
is a safe and effective treatment for
premenopausal women with hormone
receptor-positive, recurrent or advanced
breast cancer. The treatment may
become viable in the future, particularly
when tamoxifen is ineffective or contraindicated. Further studies and discussions are warranted.
Managed Care Implications: The combination of subcutaneous goserelin and
oral anastrozole may offer an alternative
to women with recurrent or metastatic
hormone receptor-positive breast cancer
prior to the initiation of intravenous
chemotherapy.
Title: Sorafenib with gemcitabine or
capecitabine in patients with HER-2
negative advanced breast cancer that
progressed during or after bevacizumab.
Authors: Schwartzberg LS, Tauer KW, Hermann RC, et al.
Reference: Clin Cancer Res. 2013;19(10);
2745-2754.
Purpose: Angiogenesis plays a critical role
in the development and local progression of breast cancer and is associated
with progression of metastatic disease.
Targeted therapies that inhibit angiogenesis have become an important
strategy for drug development. Results
with single-agent angiogenesis inhibitors in metastatic breast cancer (MBC)
have been less than optimal; however,
preclinical studies suggest the combination of such agents with chemotherapy
may be beneficial. Sorafenib (Nexavar)
is an oral small molecule inhibitor of
numerous tyrosine kinases and has
antiproliferative and antiangiogenic
activity. Information from phase 2
studies showed activity in patients with
metastatic clear cell renal cell carcinoma refractory to sunitinib (Sutent)
or bevacizumab (Avastin), meaning
that sorafenib’s activity mechanism of
action differs from those two agents.
The activity of sorafenib in combination
with agents effective in the treatment of
MBC has been undertaken. This report
assesses whether sorafenib in combination with gemcitabine (Gemzar) or
capecitabine (Xeloda) could overcome
clinical resistance to bevacizumab in
patients with MBC.
Methods: Patients ≥ 18 years of age with a
histologically or cytologically confirmed
HER2 (-) adenocarcinoma of the breast
with locally advanced (inoperable) or
metastatic disease were eligible. Other
criteria included disease progression
during or after treatment with bevacizumab in the adjuvant or metastatic
setting, an ECOG performance status
of 0 or 1, and adequate bone marrow,
hepatic and renal function. This doubleblind, randomized, placebo-controlled,
phase 2b study stratified patients in a
1:1 ratio to treatment with sorafenib
400 mg orally twice a day or placebo
and gemcitabine 1,000 mg/m2 intravenously on days one and eight repeated
every 21 days or capecitabine at a dose
of 1,000 mg/m2 orally twice a day
for the first 14 days of a 21-day cycle.
The primary end point of the study
was progression-free survival (PFS).
Secondary end points included time
to progression (TTP), overall response
rate (ORR), overall survival (OS) and
safety.
Results: A total of 160 patients
were randomized for treatment.
The majority of patients received
gemcitabine (82.5 percent, or 132
patients) with the remainder treated
with capecitabine. The vast majority of patients, 95 percent, had stage
IV disease, with 84.4 percent having
visceral disease. The combination
of sorafenib plus gemcitabine or
capecitabine provided a small but
statistically significant improvement
in PFS compared with placebo (3.4
vs. 2.7 months; p = 0.02), with a
35 percent reduction in the risk of
disease progression or death (HR
= 0.65; 95 percent CI, 0.45-0.95).
TTP was also increased (median
3.6 vs. 2.7 months; HR = 0.64; 95
percent CI, 0.44-0.93; p = 0.02).
The ORR was 19.8 vs. 12.7 percent,
again favoring sorafenib, but was
not statistically significant. Median
survival was 13.4 vs. 11.4 months for
sorafenib versus placebo (HR = 1.01;
95 percent CI, 0.71-1.44; p = 0.95).
Treatment was associated with manageable toxicity, but patients receiving
combination therapy with sorafenib
required more dose reductions when
compared to the matched placebo
controls (51.9 vs. 7.8 percent). Grade 3
toxicity occurred in 70 percent of the
patients treated with sorafenib versus
47 percent in the placebo arm. The
most frequent adverse events in the
sorafenib arm were stomatitis (10 vs. 0
percent with placebo), fatigue (18 vs. 9
percent), anemia (5 vs. 0 percent) and
hand-foot syndrome (39 vs. 5 percent).
Conclusion: The addition of sorafenib to
gemcitabine or capecitabine provided
a small but statistically significant
improvement in PFS in patients with
HER2 (-) advanced MBC whose disease had progressed on bevacizumab.
The combination therapy was associated with manageable toxicity but
required frequent reduction in dosing.
Managed Care Implications: With the
use of bevacizumab in question for
patients with MBC, oral sorafenib may
offer an alternative. Additional studies
are needed.
managedcareoncology.com
| 45
resources &
references
American Cancer Society (ACS).
The ACS is a national, community-based
volunteer health organization that offers
programs for education, patient service,
advocacy and rehabilitation. The website
includes pages on breast cancer, including
information about risk factors, diagnosis,
staging and treatment.
www.cancer.org
American Society of Clinical Oncology
(ASCO). This nonprofit organization is
committed to conquering cancer through
research, education, prevention and
delivery of high-quality care. The breast
cancer Web portal of ASCO’s official
journal, the Journal of Clinical Oncology, is
a resource for physicians to discover the
latest findings in the area of breast cancer
research through scientific abstracts, Virtual
Meeting, guidelines and other materials.
http://breast.jco.org
BreastCancer.org. This nonprofit organization is dedicated to providing information and a community for those affected
by breast cancer. Topics covered include
lowering risk, symptoms and diagnosis,
and treatment and side effects.
www.breastcancer.org
Breast Cancer Research Foundation
(BCRF). The foundation’s mission is to
achieve prevention and a cure for breast
cancer in our lifetime by providing critical
funding for innovative clinical and translational research at leading medical centers
worldwide and increasing public awareness about good breast health. The website
includes a breast health library, research
information and more.
www.bcrfcure.org
This resource guide features links and websites on breast cancer
that may be of use to the reader in daily practice.*
Living Beyond Breast Cancer. This
education and support organization
offers specialized programs to the newly
diagnosed and programs for caregivers
and health care professionals to help
them better meet the needs of women
with breast cancer. The website features
basic information about breast cancer,
news, information on clinical trials and
recommended resources.
www.lbbc.org
National Breast Cancer Coalition
(NBCC). Since its founding in 1991,
NBCC has changed the world of breast
cancer — in public policy, science,
industry and advocacy — by creating
new partnerships, collaborations,
research funding opportunities and
avenues for access to quality care. In
September 2010, NBCC launched an
initiative to end the disease by the year
2020. The website features facts and
figures, news and advocacy information.
www.breastcancerdeadline2020.org
CancerNet. This is the American Society
of Clinical Oncology’s patient information website featuring peer-reviewed
information on breast cancer, including
male, inflammatory and metaplastic
breast cancers. Statistics, staging and treatment information, medical illustrations
and research information are available.
www.cancer.net/cancer-types
MedlinePlus. A service of the U.S. Library
of Medicine and U.S. National Institutes
of Health, this website offers links to
peer-reviewed articles and abstracts on
breast cancer, clinical trial information,
glossaries, statistics and much more.
www.nlm.nih.gov/medlineplus/
breastcancer.html
*Note: Magellan Pharmacy Solutions does not endorse or verify the information presented.
46
| managedcareoncology Quarter 3 2013
National Breast Cancer Foundation
Inc. NBCF’s mission is to increase
awareness through education and
provide nurturing support services
to patients and survivors, as well as
mammograms for patients in need.
www.nationalbreastcancer.org
National Cancer Institute (NCI). The
NCI, part of the U.S. National Institutes
of Health, conducts and supports
cancer-related research, training and
health information dissemination.
This website provides information
on treatment, screening, testing and
clinical trials, research and literature
related to breast cancer.
www.cancer.gov/cancertopics/types/
breast
National Comprehensive Cancer
Network (NCCN). The NCCN
publishes clinical practice guidelines
that are developed through an
evidence-based process, including the
current practice guidelines for breast
cancer. Users must register to access
guidelines.
www.nccn.org/professionals/
physician_gls/f_guidelines.asp#site
SHARE — Self-Help for Women
with Breast or Ovarian Cancer.
SHARE supports, educates and
empowers people affected by breast
or ovarian cancer. The organization
helps people face their feelings and
fears, communicate effectively with
their physicians and make informed
decisions about their health.
www.sharecancersupport.org
Mage llan P ha rM acy So l ut io nS
Powerful Solutions, Proven Results
Magellan Pharmacy Solutions is redefining how health care manages
pharmaceutical benefit spend. Our solutions can stand alone or be integrated
into Total Drug Solutions∞, helping you manage the unseen components of
drug spend. Any pharmacy. Any benefit. Anywhere.
Our comprehensive solutions drive healthier outcomes as well as a healthy
return on your investment:
Ÿ Specialty Pharmacy
Ÿ Pharmacy Benefit Management
Ÿ Medical Pharmacy
Discover how we can partner with you to control
skyrocketing pharmaceutical costs by calling
886-664-2673. Put our innovative solutions
to work for you.
Learn more by visiting
TotalDrugSolutions.com
or snap the QR code with
your smart phone.
©2013 Amgen Inc. All rights reserved. 67426-R2-V1 5-13