08.01.2019 Views

HSS Rheumatology Annual Report 2017-2018

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

Advancing <strong>Rheumatology</strong><br />

Division of <strong>Rheumatology</strong> | <strong>Annual</strong> <strong>Report</strong> <strong>2017</strong>–<strong>2018</strong>


Table of Contents<br />

<strong>2017</strong> –<strong>2018</strong> Achievements and<br />

Fast Facts<br />

2<br />

Cutting-Edge Initiatives<br />

Identifying high-risk pregnancies<br />

in patients with lupus<br />

Improving outcomes for RA patients<br />

undergoing joint replacement<br />

Creating a better future for<br />

scleroderma patients<br />

Advancing high-value care and<br />

outcomes for patients<br />

Targeting interferon for better<br />

control of lupus<br />

Defining a new form of arthritis<br />

in cancer patients<br />

Improving bone health in patients<br />

with ankylosing spondylitis<br />

New possibilities for<br />

scleroderma treatment<br />

Using precision medicine to refine<br />

RA treatment<br />

Battling bone loss with a promising<br />

microRNA-targeting therapy<br />

Unraveling the molecular<br />

mysteries of lupus<br />

A promising new target for treating<br />

autoimmune diseases<br />

Investigating stem cells for<br />

repairing fibrotic skin<br />

Optimizing postsurgical outcomes<br />

Complex Cases<br />

Empowering Patients Through<br />

Support and Education Programs<br />

<strong>Rheumatology</strong> Leaders of the Future<br />

Notable References<br />

4<br />

5<br />

6<br />

7<br />

8<br />

9<br />

10<br />

11<br />

12<br />

13<br />

14<br />

15<br />

16<br />

17<br />

18<br />

24<br />

26<br />

31<br />

Message from the Physician-in-Chief<br />

and Chief of the Division of<br />

<strong>Rheumatology</strong><br />

<strong>HSS</strong> rheumatologists have a long history of<br />

commitment to advancing our field through<br />

our many leadership roles, our engagement<br />

in the education of future physicians, and<br />

our contributions to unraveling the complex<br />

mechanisms responsible for autoimmune and<br />

inflammatory rheumatic diseases.<br />

I want to congratulate Anne R. Bass, MD,<br />

Attending Physician, on completing her term as<br />

Chair of the American College of <strong>Rheumatology</strong>’s<br />

(ACR) Committee on <strong>Rheumatology</strong> Training and<br />

Workforce Issues. In view of the acknowledged<br />

need to grow the number of adult rheumatologists<br />

and pediatric rheumatologists in the U.S., recruiting<br />

and training physicians is a high priority nationally<br />

and at <strong>HSS</strong>. Dr. Bass has done an outstanding<br />

job in her ACR role and as Director of the <strong>HSS</strong><br />

<strong>Rheumatology</strong> Training Program.<br />

<strong>HSS</strong> rheumatologists also have high visibility and<br />

critical roles in the programs at our academic<br />

partner institution, Weill Cornell Medicine. Jane<br />

E. Salmon, MD, Attending Physician, serves as<br />

Associate Dean for Faculty Affairs. Juliet B. Aizer,<br />

MD, Assistant Attending Physician, and Edward J.<br />

Parrish, MD, Assistant Attending Physician, have<br />

designed significant portions of the curriculum<br />

for first- and second-year medical students and<br />

guide nearly all members of our faculty in teaching<br />

activities at the medical school.<br />

“<strong>HSS</strong> rheumatologists and scientists are revolutionizing the care of patients<br />

with some of the most challenging, complex medical conditions through<br />

transformative research and global professional education.”<br />

— Louis A. Shapiro, President and Chief Executive Officer, and Todd J. Albert, MD, FACS,<br />

Surgeon-in-Chief and Medical Director, Korein-Wilson Professor of Orthopaedic Surgery<br />

On cover, top left: From Nathalie Burg, MD, Instructor. Lower right: From Shipman et al., “A protective Langerhans cell-keratinocyte axis that is<br />

dysfunctional in photosensitivity.” Science Translational Medicine 15 Aug <strong>2018</strong>: Vol. 10, Issue 454, eaap9527. Reprinted with permission from AAAS.


Perhaps only second to our focus on expert<br />

patient care is our commitment to studying the<br />

mechanisms of lupus, scleroderma, myositis and<br />

other rheumatic diseases: Why do they occur? Why<br />

do they affect some people and not others? Why<br />

are they more common in women than in men?<br />

<strong>HSS</strong> has a long history of supporting physicianscientists.<br />

Over the last five years, we’ve recruited<br />

seven junior faculty members who are pursuing<br />

careers in clinical and translational research. We<br />

also founded the <strong>HSS</strong> <strong>Rheumatology</strong> Council, a<br />

committee of supporters and donors dedicated to<br />

raising the <strong>Rheumatology</strong> Division’s visibility and<br />

enabling research.<br />

Working in partnership with the Development<br />

Department, we initiated a research grant program<br />

and were pleased to award the first <strong>Rheumatology</strong><br />

Council Grants to the following physicians:<br />

• Karmela K. Chan, MD,<br />

Assistant Attending Physician<br />

• Jessica K. Gordon, MD,<br />

Assistant Attending Physician<br />

• Nathalie Burg, MD, Instructor<br />

• David R. Fernandez, MD, PhD,<br />

Assistant Attending Physician<br />

Other grant recipients include Bella Mehta,<br />

MBBS, MD, Assistant Attending Physician,<br />

who received the C. Ronald MacKenzie Young<br />

Scientist Endowment Award. Michela Manni, PhD,<br />

Instructor, received the Lockshin Fellowship Award<br />

from the Barbara Volcker Center for Women and<br />

Rheumatic Disease.<br />

Our five Centers of Excellence and Division of<br />

Pediatric <strong>Rheumatology</strong> facilitate collaboration<br />

among physicians with disease-related academic<br />

interests. Clinical care, research and patient<br />

support are organized through most of the<br />

Centers. Dr. Salmon, Director of the Lupus and<br />

Antiphospholipid Syndrome Center of Excellence,<br />

and colleagues oversee the recruitment of<br />

patients for translational research studies on<br />

lupus and APS and prioritize interventional clinical<br />

trials of candidate therapeutics. The Center also<br />

works closely with <strong>HSS</strong> social workers to lead<br />

support groups and educational programs for<br />

patients. <strong>HSS</strong> has been dedicated to advancing<br />

the understanding of the immunopathogenesis<br />

of systemic lupus erythematosus for many years,<br />

and we continue to identify biomarkers that<br />

predict complications of disease and molecular<br />

targets that hold promise of leading to new<br />

drug therapies.<br />

Dr. Gordon and Robert F. Spiera, MD, Director of<br />

the Scleroderma, Vasculitis & Myositis Center of<br />

Excellence and Attending Physician, have worked<br />

closely with Franck Barrat, PhD, Senior Scientist<br />

at the <strong>HSS</strong> Research Institute, to study the role of<br />

plasmacytoid dendritic cells in systemic sclerosis.<br />

They have identified Toll-like receptor 8 as a novel<br />

pathway for immune system activation in patients<br />

with the disease. Drs. Gordon and Spiera are also<br />

designing investigator-initiated studies of novel<br />

therapeutics and participating in multicenter<br />

sponsored clinical trials.<br />

<strong>HSS</strong> rheumatologists recognize the value of<br />

close collaboration with basic scientists in the<br />

laboratory as well as with orthopaedic surgeons.<br />

With the rich patient resources available at<br />

<strong>HSS</strong>, clinical data and synovial tissue samples<br />

collected at the time of arthroplasty have been<br />

studied to characterize the cell populations<br />

that are associated with flares in rheumatoid<br />

arthritis. Vivian P. Bykerk, MD, Director of the<br />

Inflammatory Arthritis Center of Excellence<br />

and Associate Attending Physician, and Susan<br />

M. Goodman, MD, Director of the Integrative<br />

<strong>Rheumatology</strong> and Orthopedic Center of<br />

Excellence and Attending Physician, have<br />

established protocols that provide the clinical<br />

data necessary for productive analysis of<br />

tissue samples. These researchers and their<br />

collaborators are using machine learning to<br />

analyze gene expression data from synovial<br />

tissue cells in relation to pathologic patterns of<br />

tissue histology in rheumatoid arthritis patients.<br />

In our Bone Health and Osteoporosis Center<br />

of Excellence, Emily M. Stein, MD, Associate<br />

Attending Physician and Associate Scientist, is<br />

working closely with the <strong>HSS</strong> Spine Service to<br />

study bone quality.<br />

Our commitment to young investigators and the<br />

success of their academic careers will ensure<br />

that we sustain our long tradition of leadership in<br />

rheumatology and our history of advancing patient<br />

care. Our application of current technologies<br />

to characterize the molecular mechanisms that<br />

underlie the diseases we treat exemplifies our<br />

unwavering commitment to achieve better lives for<br />

our patients.<br />

Mary K. Crow, MD<br />

Physician-in-Chief<br />

Chair, Department of Medicine<br />

Chief, Division of <strong>Rheumatology</strong><br />

Benjamin M. Rosen Chair in Immunology and<br />

Inflammation Research<br />

Joseph P. Routh Professor of Rheumatic Disease Medicine


2<br />

Division of <strong>Rheumatology</strong><br />

<strong>2017</strong>–<strong>2018</strong> Achievements<br />

#3 in the nation<br />

for <strong>Rheumatology</strong> by U.S. News & World <strong>Report</strong> “Best Hospitals”<br />

(<strong>2018</strong>–2019 rankings)<br />

The Division of <strong>Rheumatology</strong>, the Department of Social<br />

Work Programs and the Digital Communications team created<br />

LupusMinder, a smartphone app that helps lupus patients manage<br />

their condition. In <strong>2018</strong>, the app received a Platinum award in<br />

the category of Best Native Mobile App from the eHealthcare<br />

Leadership Awards.<br />

Mary K. Crow, MD, Physician-in-Chief and Chief, Division<br />

of <strong>Rheumatology</strong>, received the Presidential Gold Medal<br />

from the American College of <strong>Rheumatology</strong> (ACR)<br />

in <strong>2018</strong>. It is awarded in recognition of outstanding<br />

achievements in rheumatology over the course of a career.<br />

Dr. Crow is Co-Chair of the Scientific Advisory Board<br />

of the Lupus Research Alliance and a member of the<br />

Executive Committee of the Lupus Clinical Investigators<br />

Network (LuCIN).<br />

Franck Barrat, PhD, Senior Scientist; Robert F. Spiera,<br />

MD, Director of the Scleroderma, Vasculitis & Myositis<br />

Center of Excellence and Attending Physician; Jessica<br />

K. Gordon, MD, Assistant Attending Physician; David<br />

R. Fernandez, MD, Assistant Attending Physician; and<br />

other <strong>HSS</strong> investigators published a significant study<br />

in Science Translational Medicine in <strong>2018</strong> identifying<br />

the plasmacytoid dendritic cell and Toll-like receptor<br />

8 as important contributors to the pathogenesis of<br />

systemic sclerosis.<br />

Anne R. Bass, MD, Attending Physician, served as Chair<br />

of the ACR Committee on <strong>Rheumatology</strong> Training and<br />

Workforce Issues.<br />

Vivian P. Bykerk, MD, Director of the Inflammatory<br />

Arthritis Center of Excellence and Associate Attending<br />

Physician, is the lead investigator of a rheumatoid arthritis<br />

(RA) study funded by the National Institutes of Health<br />

(NIH) and partnering pharmaceutical companies as part<br />

of the Accelerating Medicines Partnership (AMP). The<br />

study is focused on identifying molecular processes in<br />

synovial tissue that reflect disease flare.<br />

Doruk Erkan, MD, MPH, Associate Attending Physician and<br />

Clinical Co-Director of the Mary Kirkland Center for Lupus<br />

Care, is Executive Committee Chair of the international APS<br />

research network APS ACTION (Antiphospholipid Syndrome<br />

Alliance for Clinical Trials and International Networking).<br />

Susan M. Goodman, MD, Director of the Integrative<br />

<strong>Rheumatology</strong> and Orthopedic Center of Excellence and<br />

Attending Physician, led the expert panel that created the<br />

first-ever Guideline for the Perioperative Management<br />

of Antirheumatic Medication in Patients With Rheumatic<br />

Diseases Undergoing Elective Total Hip or Total Knee<br />

Arthroplasty. The Guideline is sponsored by the American<br />

College of <strong>Rheumatology</strong> and the American Association of<br />

Hip and Knee Surgeons.<br />

Kyriakos A. Kirou, MD, DSc, Assistant Attending<br />

Physician, is the <strong>HSS</strong> Site Director of LuCIN.<br />

Michael D. Lockshin, MD, Director of the Barbara<br />

Volcker Center for Women and Rheumatic Disease and<br />

Attending Physician, is author of The Prince at the Ruined<br />

Tower: Time, Uncertainty & Chronic Illness, which was<br />

published in <strong>2017</strong>.<br />

Dr. Lockshin and Dr. Erkan are editors of<br />

Antiphospholipid Syndrome: Current Research Highlights<br />

and Clinical Insights, which was published in <strong>2017</strong>.<br />

Theresa T. Lu, MD, PhD, Associate Scientist, was<br />

awarded a research grant from the Lupus Research<br />

Alliance to study the involvement of the lymphatic<br />

system in ultraviolet light-induced cutaneous lupus.<br />

Catherine H. MacLean, MD, PhD, Chief Value<br />

Medical Officer and Director of the <strong>HSS</strong> Center for the<br />

Advancement of Value in Musculoskeletal Care, was<br />

named in the inaugural “Notable Women in Health Care<br />

in NYC” list in Crain’s New York Business. In addition,<br />

she serves on and is the immediate past chair of the<br />

performance measures committee for the American<br />

College of Physicians.<br />

Bella Mehta, MBBS, MD, Assistant Attending Physician,<br />

received the ACR Distinguished Fellow Award in <strong>2018</strong>.


Hospital for Special Surgery 3<br />

Karen B. Onel, MD, Chief of the Division of Pediatric<br />

<strong>Rheumatology</strong> and Attending Physician, is Chair of the<br />

Childhood Arthritis and <strong>Rheumatology</strong> Research Alliance<br />

(CARRA) Ethics Committee. She is Co-Chair of the CARRA<br />

Systemic Juvenile Idiopathic Arthritis working group.<br />

Jillian Rose, LCSW, MPH, Director of Community<br />

Engagement, Diversity and Research, received the<br />

Distinguished Educator Award from the Association of<br />

<strong>Rheumatology</strong> Health Professionals in <strong>2018</strong>.<br />

Linda A. Russell, MD, Director of the Bone Health<br />

and Osteoporosis Center of Excellence and Associate<br />

Attending Physician, was appointed by the American<br />

College of <strong>Rheumatology</strong> as a member of its Committee<br />

on Rheumatologic Care.<br />

Jane E. Salmon, MD, Director of the Lupus and<br />

Antiphospholipid Syndrome Center of Excellence and<br />

Attending Physician, and Dr. Crow were named Honorary<br />

Members of the European League Against Rheumatism<br />

(EULAR) in <strong>2018</strong> and <strong>2017</strong>, respectively. They are among<br />

the first four Americans to achieve this honor, which<br />

recognizes their contributions to research, education and<br />

service to EULAR.<br />

Dr. Salmon is a member of the Steering Committee of<br />

LuCIN and the Scientific Advisory Board of the Lupus<br />

Research Alliance. She was named a Master of the<br />

American College of <strong>Rheumatology</strong> in <strong>2017</strong>.<br />

Lisa R. Sammaritano, MD, Associate Attending<br />

Physician, was Principal Investigator of the American<br />

College of <strong>Rheumatology</strong> Reproductive Health in<br />

Rheumatic Diseases Guideline.<br />

Robert F. Spiera, MD, Director of the Scleroderma,<br />

Vasculitis & Myositis Center of Excellence and Attending<br />

Physician, was a member of a group of investigators that<br />

designed and executed a clinical trial for a new therapy<br />

for giant-cell arteritis. Tocilizumab, an interleukin (IL)-6<br />

blocking agent, was the first therapy approved by the<br />

U.S. Food & Drug Administration and European Medicines<br />

Agency for this indication.<br />

Dr. Spiera is Chair of the Medical and Scientific Advisory<br />

Board of the Scleroderma Foundation, Tri-State Chapter.<br />

He and Jessica K. Gordon, MD, Assistant Attending<br />

Physician, are members of this Board.<br />

Dr. Spiera and Dr. Gordon are members of the National<br />

Medical and Scientific Advisory Board of the Scleroderma<br />

Foundation.<br />

Sarah F. Taber, MD, Assistant Attending Physician,<br />

is a co-author of the chapter on Systemic Lupus<br />

Erythematosus in Childhood and Adolescence in the<br />

<strong>2018</strong> edition of Dubois’ Lupus Erythematosus and<br />

Related Syndromes.<br />

Fast Facts<br />

Division of <strong>Rheumatology</strong><br />

The Division of <strong>Rheumatology</strong> comprises five Centers of Excellence and the Division of Pediatric <strong>Rheumatology</strong>.<br />

The Centers bring together <strong>HSS</strong> faculty and staff members with an interest in a particular disease area to<br />

develop projects and new research initiatives. They include physicians and healthcare professionals from other<br />

departments, such as Orthopaedics, Social Work, Nursing and Research.<br />

• Bone Health and Osteoporosis Center of Excellence<br />

• Inflammatory Arthritis Center of Excellence<br />

• Integrative <strong>Rheumatology</strong> and Orthopedic Center of Excellence<br />

• Lupus and Antiphospholipid Syndrome Center of Excellence<br />

• Scleroderma, Vasculitis & Myositis Center of Excellence<br />

• Division of Pediatric <strong>Rheumatology</strong><br />

Adult rheumatologists<br />

36 36,027<br />

Pediatric rheumatologists<br />

5 2,969<br />

Scientists<br />

9 2,728<br />

Perioperative<br />

17 medicine physicians 15,665<br />

9<br />

<strong>Rheumatology</strong><br />

support and<br />

education programs for patients<br />

Adult rheumatology<br />

patient visits<br />

Pediatric rheumatology<br />

patient visits<br />

Endocrinology<br />

patient visits<br />

Perioperative<br />

patient visits<br />

All figures pertain to the <strong>2017</strong>–<strong>2018</strong> academic year.


4<br />

Division of <strong>Rheumatology</strong><br />

Identifying high-risk pregnancies<br />

in patients with lupus<br />

Most women with lupus can<br />

have successful pregnancies,<br />

but some develop complications,<br />

such as preeclampsia, fetal<br />

and neonatal death, and<br />

fetal growth restriction. We<br />

cannot predict in whom these<br />

adverse outcomes will occur.<br />

In experimental laboratory<br />

models, complement activation<br />

causes adverse pregnancy<br />

outcomes. To investigate the<br />

role of complement in patients,<br />

Jane E. Salmon, MD, Collette<br />

Kean Research Chair, Attending<br />

Physician and Director of the<br />

Lupus and Antiphospholipid<br />

Syndrome Center of Excellence;<br />

Michael D. Lockshin, MD, Director<br />

of the Barbara Volcker Center<br />

for Women and Rheumatic<br />

Disease and Attending Physician;<br />

and Lisa R. Sammaritano, MD,<br />

Associate Attending Physician,<br />

and colleagues mined the data<br />

and samples from PROMISSE<br />

(Predictors of Pregnancy<br />

Outcome: Biomarkers in<br />

Antiphospholipid Antibody<br />

Syndrome and Systemic Lupus<br />

Erythematosus). The results of<br />

their study, which was published<br />

in the Annals of the Rheumatic<br />

Diseases in April <strong>2018</strong>, included<br />

the evaluation of 487 pregnant<br />

women with SLE and/or aPL<br />

antibodies and 204 pregnant<br />

healthy controls. At each<br />

month of pregnancy, samples<br />

were obtained to measure<br />

complement activation products<br />

in patients’ blood. Adverse<br />

pregnancy outcomes occurred<br />

in approximately 20 percent of<br />

SLE and/or aPL pregnancies<br />

and were associated with<br />

lupus anticoagulant, history of<br />

hypertension, and non-white<br />

race. Levels of complement<br />

products early in pregnancy<br />

were significantly higher in<br />

A recent finding by Jane E. Salmon, MD, Director of the Lupus and<br />

Antiphospholipid Syndrome Center of Excellence and Attending Physician<br />

(left), may help pregnant women with lupus have healthy babies, such as<br />

the patient shown at right. Marta M. Guerra, MS, Project Coordinator for<br />

Dr. Salmon’s study (center), admires the baby.<br />

patients who experienced<br />

pregnancy complications.<br />

These findings suggest that,<br />

as in the experimental models,<br />

complement activation<br />

contributes to abnormal placental<br />

development, which leads to<br />

pregnancy complications.<br />

TNF-α is released by<br />

inflammatory cells as a<br />

consequence of complement<br />

activation, and it too contributes<br />

to placental dysfunction,<br />

preeclampsia and fetal growth<br />

restriction in experimental<br />

models. To translate these<br />

findings to patients, Dr. Salmon<br />

and her team are conducting<br />

a trial in which they will treat<br />

pregnant women with APS and<br />

SLE at high risk for serious<br />

pregnancy complications with<br />

certolizumab, a TNF inhibitor used<br />

Dr. Salmon’s<br />

research suggests<br />

that complement<br />

activation contributes<br />

to abnormal placental<br />

development, which<br />

leads to pregnancy<br />

complications.<br />

to treat rheumatoid arthritis and<br />

psoriatic arthritis that does not<br />

cross the placenta. “If the trial<br />

prevents pregnancy complications<br />

in patients with SLE and APS, it<br />

may also prevent preeclampsia<br />

in women without autoimmune<br />

disease who are at risk for<br />

placental insufficiency,” says<br />

Dr. Salmon. “We hope our studies<br />

of SLE and APS have implications<br />

for the general population.”


Hospital for Special Surgery 5<br />

Improving outcomes for<br />

RA patients undergoing<br />

joint replacement<br />

Although rheumatoid arthritis<br />

(RA) patients have experienced<br />

improvements in healthrelated<br />

quality of life and<br />

functional status, rates of total<br />

hip replacement and total<br />

knee replacement procedures<br />

have remained stable. Most<br />

RA patients who undergo<br />

arthroplasty are taking diseasemodifying<br />

antirheumatic drugs<br />

and biologic therapies, which<br />

are known to increase their risk<br />

of developing a prosthetic joint<br />

infection. As a result, patients<br />

are typically instructed to stop<br />

taking their medications prior<br />

to surgery. This may increase<br />

their risk of having a flare<br />

perioperatively, which may have<br />

a negative effect on rehabilitation<br />

efforts and outcomes.<br />

To investigate patient-reported<br />

RA flares, <strong>HSS</strong> researchers<br />

launched the RA Perioperative<br />

Flare Study. This is likely the<br />

first study to prospectively<br />

assess postoperative, patientreported<br />

flares of RA in patients<br />

undergoing THA and TKA using<br />

the RA-FQ, a validated, patientreported<br />

outcome instrument for<br />

identifying RA flares.<br />

In the study, which was published<br />

in The Journal of <strong>Rheumatology</strong><br />

in May <strong>2018</strong>, 120 <strong>HSS</strong> patients<br />

answered a questionnaire each<br />

week for six consecutive weeks<br />

after surgery. Patients who<br />

reported flares after surgery were<br />

compared to those who did not<br />

experience them. Lead author<br />

Susan M. Goodman, MD, Director<br />

of the Integrative <strong>Rheumatology</strong><br />

and Orthopedic Center of<br />

Excellence and Attending<br />

From left to right: Mark P. Figgie, MD, Chief of the Surgical Arthritis Service,<br />

Allan E. Inglis MD Chair in Surgical Arthritis, and Attending Orthopaedic<br />

Surgeon, and Susan M. Goodman, MD, Director of the Integrative<br />

<strong>Rheumatology</strong> and Orthopedic Center of Excellence and Attending<br />

Physician, are investigating flares in RA patients after arthroplasty.<br />

Physician, and colleagues found<br />

that 63 percent of the patients<br />

experienced a flare by six weeks<br />

after surgery. The patients who<br />

flared had significantly higher<br />

disease activity at baseline, but<br />

preoperative antirheumatic<br />

medication withdrawal was not an<br />

independent risk factor for flares.<br />

To improve outcomes and<br />

reduce the risk of infection in<br />

RA patients, <strong>HSS</strong> researchers<br />

are investigating the effects<br />

of changing the perioperative<br />

medication protocol. “We can<br />

withhold biologic medications<br />

for one-dose intervals around<br />

the time of surgery,” says<br />

Dr. Goodman. “Now we’re looking<br />

at pain and functionality in these<br />

patients after a year.”<br />

Rheumatoid arthritis<br />

patients who<br />

experienced flares<br />

after arthroplasty<br />

had significantly<br />

higher disease<br />

activity at baseline.


6<br />

Division of <strong>Rheumatology</strong><br />

Creating a better future for<br />

scleroderma patients<br />

Scleroderma is one of the<br />

most debilitating rheumatic<br />

diseases. It is associated with<br />

significant morbidity and<br />

disability and has the highest<br />

rate of all-cause mortality<br />

among the rheumatic diseases.<br />

To date, no medication has<br />

been specifically approved for<br />

this condition. Fortunately,<br />

<strong>HSS</strong> physicians and scientists<br />

have made significant progress<br />

in investigating promising<br />

new treatment options. One<br />

area of focus is lenabasum,<br />

a cannabinoid receptor<br />

type 2 (CB2) agonist. In<br />

<strong>2017</strong>, Robert F. Spiera, MD,<br />

Director of the Scleroderma,<br />

Vasculitis & Myositis Center<br />

of Excellence and Attending<br />

Physician, led a Phase II<br />

study of the drug sponsored<br />

by Corbus Pharmaceuticals.<br />

It demonstrated acceptable<br />

safety and tolerability and<br />

suggested a possible benefit<br />

in scleroderma patients. He<br />

presented the results at the<br />

<strong>2017</strong> American College of<br />

<strong>Rheumatology</strong> <strong>Annual</strong> Meeting.<br />

CB2 receptors are expressed on<br />

immune system cells, including<br />

T cells, macrophages and B<br />

cells. They appear to modulate<br />

immune function. “Triggering<br />

these cells turns off the<br />

propagation of the inflammation<br />

phase of the innate immune<br />

response and promotes<br />

expression of resolvins, which<br />

contribute to resolving the<br />

inflammatory response, thereby<br />

ultimately diminishing tissue<br />

fibrosis,” says Dr. Spiera.<br />

In a one-year, open label<br />

extension study that followed<br />

the Phase II trial, patients<br />

taking lenabasum continued to<br />

see significant improvements<br />

in inflammation and fibrosis.<br />

From left to right: Jessica K. Gordon, MD, Assistant Attending Physician,<br />

and Robert F. Spiera, MD, Director of the Scleroderma, Vasculitis & Myositis<br />

Center of Excellence and Attending Physician, are studying two drugs that<br />

may be effective in scleroderma patients.<br />

Dr. Spiera presented these<br />

results at the European League<br />

Against Rheumatism Congress<br />

in Amsterdam in June <strong>2018</strong>. Now<br />

<strong>HSS</strong> is the lead site for a yearlong<br />

Phase III trial of lenabasum,<br />

which is currently enrolling<br />

patients around the world.<br />

Positive results could help to<br />

advance the drug on the path to<br />

FDA approval. “What’s appealing<br />

about this drug is it appears to<br />

be very safe,” says Dr. Spiera.<br />

Unlike medications that<br />

suppress the immune system,<br />

lenabasum doesn’t appear to<br />

increase the risk of infection.<br />

Dr. Spiera is also optimistic<br />

about belimumab. In an <strong>HSS</strong>sponsored,<br />

investigator-initiated<br />

trial of the drug, patients with<br />

early diffuse scleroderma who<br />

received the medication saw<br />

significant improvements in skin<br />

thickness scores. The findings<br />

were published in Arthritis and<br />

<strong>Rheumatology</strong> in January <strong>2018</strong>.<br />

The researchers also<br />

determined that the drug<br />

affects patients’ gene<br />

expression in skin biopsies.<br />

“Significant decreases in the<br />

expression of B-cell signaling<br />

and pro-fibrotic gene pathways<br />

were observed in patients who<br />

improved on the drug and not<br />

in those who didn’t improve,”<br />

says Dr. Spiera. This supports<br />

the idea that the drug was<br />

having an effect on the relevant<br />

cellular mechanisms. Dr. Spiera<br />

and his colleague, Jessica<br />

K. Gordon, MD, Assistant<br />

Attending Physician, plan to<br />

initiate a larger study of this<br />

strategy, which will begin in<br />

early 2019.<br />

Although there’s a long road<br />

ahead for getting a new drug<br />

to market, the prospects are<br />

promising. “It will be very<br />

exciting if either drug pans<br />

out,” says Dr. Spiera. “This is a<br />

totally unmet need.”


Hospital for Special Surgery 7<br />

Advancing high-value care<br />

and outcomes for patients<br />

Catherine H. MacLean, MD, PhD, Chief Value Medical Officer, is studying<br />

patient-reported outcome measures, or PROMs.<br />

Our commitment to providing<br />

value-based care is reflected in<br />

our high-quality services and<br />

low complication rates. “<strong>HSS</strong><br />

leads the country in performance<br />

on a number of different safety<br />

measures,” says Catherine H.<br />

MacLean, MD, PhD, Chief Value<br />

Medical Officer. “As a result,<br />

our total cost for an episode of<br />

care is lower than that of other<br />

hospitals.” While safety is of<br />

paramount importance, patients<br />

come to <strong>HSS</strong> to get better. So we<br />

are focused on studying patientreported<br />

outcome measures,<br />

or PROMs. “We’re measuring<br />

PROMs as a standard of care<br />

because we want to understand<br />

how our patients are doing in<br />

terms of improving their pain<br />

and functional status,” says<br />

Dr. MacLean.<br />

<strong>HSS</strong> physicians also aim to be<br />

responsible when prescribing<br />

medications. “Rheumatologists<br />

are the stewards of a number of<br />

very expensive drugs, including<br />

adalimumab, infliximab,<br />

etanercept and rituximab—four<br />

of the five drugs with the highest<br />

dollar sales worldwide,” says<br />

Dr. MacLean. “These medications<br />

are often effective for rheumatoid<br />

arthritis (RA) patients, but there<br />

are less expensive alternatives<br />

that may be appropriate for<br />

certain patients. By routinely<br />

assessing disease activity and<br />

patient outcomes, we can be<br />

more thoughtful about the drugs<br />

we prescribe.” At the same time,<br />

physicians need to have open,<br />

honest conversations with their<br />

patients about the costs of<br />

medical care—in particular, these<br />

very expensive medications,<br />

which cost thousands of dollars<br />

per month. Given the choice<br />

between two similarly effective<br />

drugs, patients might prefer the<br />

less expensive option, especially<br />

if they have a high co-pay or<br />

no insurance.<br />

The highest value care for<br />

rheumatoid arthritis is that which<br />

prevents long-term disease or<br />

joint damage. Hence, our first<br />

objective is to treat early and<br />

aggressively with the best drug<br />

for the individual patient, be that<br />

a conventional drug or a more<br />

expensive biologic one. This has<br />

resulted in a marked reduction in<br />

the development of joint damage,<br />

deformity and disability. “When<br />

I was a fellow, there were always<br />

wheelchairs in the rheumatology<br />

waiting room,” says Dr. MacLean.<br />

“They are a pretty rare sight<br />

these days.”<br />

How a rheumatologist became Chief Value Medical Officer<br />

Catherine H. MacLean, MD, PhD, is a<br />

rheumatologist as well as Chief Value<br />

Medical Officer. She currently directs the<br />

<strong>HSS</strong> Center for the Advancement of Value<br />

in Musculoskeletal Care. In this role, Dr.<br />

MacLean develops programs, systems<br />

and tools to deliver high-value care at<br />

<strong>HSS</strong> and beyond. She has had a full and<br />

varied career, with stints in academic<br />

medicine, research and the insurance<br />

industry. After attending Washington<br />

University School of Medicine in St.<br />

Louis, Dr. MacLean completed training<br />

in internal medicine at Harbor-UCLA<br />

Medical Center and rheumatology at<br />

UCLA Center for the Health Sciences. As<br />

part of UCLA’s first Scientific Training and<br />

Advanced Research fellowship class, she<br />

also earned a PhD in health services from<br />

UCLA’s School of Public Health. Upon<br />

completing her training, she joined the<br />

faculty at UCLA with joint appointments<br />

at the RAND Corporation and the West<br />

Los Angeles VAMC, where her research<br />

focused on defining, measuring and<br />

improving healthcare quality. She was<br />

recruited by Anthem/WellPoint to help<br />

drive quality improvement and eventually<br />

led quality and care management<br />

programs there. In 2015, she joined <strong>HSS</strong>.<br />

Dr. MacLean was the principal<br />

investigator on many academic research<br />

projects at UCLA and RAND, and has<br />

been a director, chair or participant on<br />

numerous national committees and<br />

panels related to healthcare quality and<br />

value. She is the immediate past chair of<br />

the performance measures committee at<br />

the American College of Physicians (ACP)<br />

and currently serves on the ACP’s Quality<br />

Improvement Learning Collaborative<br />

Task Force and the National Committee<br />

for Quality Assurance’s Bone and Joint<br />

Measurement Advisory Panel.


8<br />

Division of <strong>Rheumatology</strong><br />

Targeting interferon for<br />

better control of lupus<br />

<strong>HSS</strong> has been a leader in the<br />

quest to better understand<br />

systemic lupus erythematosus<br />

(SLE), thanks in large part to<br />

research directed by Mary K.<br />

Crow, MD, Physician-in-Chief<br />

and Chief of the Division of<br />

<strong>Rheumatology</strong>. About 17 years<br />

ago, she and two other groups<br />

observed that type I interferon<br />

was responsible for the genes<br />

expressed in blood from lupus<br />

patients. This suggested that<br />

a virus or virus-like stimulus<br />

might play a role in the wide<br />

range of immune alterations and<br />

autoimmunity associated with<br />

SLE. This finding became known<br />

as the “interferon signature” and<br />

continues to be an important<br />

way of characterizing lupus<br />

patients for research.<br />

Over the last decade, Dr. Crow<br />

and her team identified an<br />

association between a positive<br />

interferon signature and<br />

active and severe disease.<br />

They also found that the<br />

interferon signature was<br />

associated with the presence<br />

of autoantibodies that target<br />

RNA or RNA-binding proteins.<br />

That observation indicated that<br />

immune complexes containing<br />

RNA, as well as RNA within<br />

cells, might be triggers for the<br />

production of type I interferon.<br />

This was confirmed in other<br />

studies. “Our lab has been<br />

particularly interested in the<br />

potential role of RNA encoded by<br />

endogenous virus-like elements<br />

in our own genomes—LINE-1<br />

retrotransposons—as drivers<br />

of type I interferon in SLE,”<br />

says Dr. Crow. “We showed<br />

that cells from patients with<br />

SLE or Sjogren’s syndrome<br />

express elevated RNAs encoding<br />

LINE‐1 and that LINE-1 RNA can<br />

induce the production of type I<br />

interferon. Overall, we think that<br />

type I interferon is responsible<br />

for many of the immune system<br />

alterations in SLE patients.”<br />

Currently, Dr. Crow is studying<br />

agents aimed at reducing the<br />

interferon signature—and the<br />

production or action of type I<br />

interferon. “We are continuing<br />

to investigate the role of<br />

endogenous retroelements<br />

as drivers of type I interferon<br />

production, but we have<br />

Left: SLE is characterized by<br />

sustained activation of the type I<br />

interferon pathway, in contrast to<br />

the brief expression of interferon<br />

after a viral infection.<br />

Above, top: Mary K. Crow, MD, Physician-in-Chief and Chief of the Division<br />

of <strong>Rheumatology</strong> (center), and her research team discuss their findings on<br />

the role of type I interferon in systemic lupus erythematosus.<br />

expanded the scope of our<br />

research to include additional<br />

molecular pathways involved<br />

in SLE,” says Dr. Crow. “We<br />

are currently analyzing gene<br />

expression data that we think<br />

will provide new insights into<br />

mechanisms responsible for<br />

lupus nephritis, an important<br />

organ system manifestation<br />

of SLE and a reason for poor<br />

patient outcomes.”


Hospital for Special Surgery 9<br />

Defining a new form of<br />

arthritis in cancer patients<br />

“Studying a new form<br />

of arthritis linked to<br />

immunotherapy drugs<br />

may help us to figure<br />

out the immunologic<br />

pathways that lead<br />

to arthritis. That may<br />

allow us to target those<br />

pathways early on and<br />

potentially prevent RA in<br />

the general population.”<br />

Anne R. Bass, MD,<br />

Attending Physician<br />

Anne R. Bass, MD, Attending Physician, examines a patient who is taking<br />

immunotherapy medications.<br />

Immunotherapy drugs known<br />

as “checkpoint inhibitors” can<br />

have dramatic, often lifesaving<br />

benefits for cancer patients.<br />

But they are also known to<br />

have problematic side effects.<br />

“Patients are staying on these<br />

drugs for longer periods of<br />

time, and we’re starting to see<br />

arthritis as a side effect more<br />

commonly,” says Anne R. Bass,<br />

MD, Attending Physician. “My<br />

theory is that some patients<br />

are genetically predisposed to<br />

arthritis and develop it when<br />

they take these immunestimulating<br />

therapies.”<br />

Collaborating with oncologists<br />

at a major New York City<br />

cancer center, Dr. Bass has<br />

been working to clinically<br />

define this new form of<br />

inflammatory arthritis,<br />

which causes joint swelling<br />

and pain and occasionally is<br />

associated with rheumatoid<br />

arthritis (RA) markers in<br />

the blood. In some patients,<br />

symptoms resemble RA, and<br />

in others, they are similar<br />

to polymyalgia rheumatica.<br />

Dr. Bass is investigating<br />

why and how the condition<br />

develops on an immunological<br />

level and is establishing<br />

treatment protocols. One key<br />

question: Is the “RA-like” form<br />

of inflammatory arthritis the<br />

same as the RA that develops<br />

in people who haven’t taken<br />

immunotherapy drugs? “If it<br />

is, then these patients offer<br />

an opportunity to study RA<br />

developing over a compressed<br />

period of time,” says Dr. Bass.<br />

This shortened timeline “may<br />

enable us to identify predictors<br />

for RA and figure out the<br />

immunologic pathways that<br />

lead to arthritis. That may allow<br />

us to target those pathways<br />

early on and potentially prevent<br />

RA in the general population.”<br />

While we aim to treat the<br />

arthritis that develops in<br />

cancer patients who are taking<br />

immunotherapy medications, we<br />

don’t want to interfere with the<br />

curative effects of the drugs by<br />

suppressing the immune system<br />

too much. Says Dr. Bass: “This<br />

is an important opportunity<br />

for clinical and research<br />

collaboration with our oncology<br />

colleagues, especially now that<br />

immunotherapy is being used<br />

for an ever-expanding array<br />

of cancers.”


10<br />

Division of <strong>Rheumatology</strong><br />

Susan M. Goodman, MD, Director of the Integrative <strong>Rheumatology</strong> and Orthopedic Center of Excellence and<br />

Attending Physician, and <strong>HSS</strong> colleagues are studying the effects of the drug secukinumab on the skeleton.<br />

Improving bone health in patients<br />

with ankylosing spondylitis<br />

While people with ankylosing<br />

spondylitis (AS) tend to<br />

experience overactive bone<br />

growth and develop abnormal<br />

bone spurs, they are also prone<br />

to osteoporosis. Studies have<br />

estimated that the prevalence<br />

of osteoporosis is as high as<br />

25 percent for these patients—<br />

more than double the 10 percent<br />

prevalence in the general<br />

population. Although these<br />

patients produce more cortical<br />

bone, the trabecular bone is<br />

abnormally weak. As a result,<br />

they are at risk of spinal fractures,<br />

which can lead to changes in<br />

posture and pain.<br />

Bisphosphonates, calcium and<br />

vitamin D can be prescribed<br />

to prevent fractures, but AS<br />

patients don’t respond well<br />

to these therapies. In <strong>2018</strong>,<br />

<strong>HSS</strong> physicians launched a<br />

pilot study to investigate the<br />

effects of secukinumab—a<br />

monoclonal antibody used to<br />

treat autoimmune diseases—<br />

on the skeleton. The drug is<br />

currently approved for treatment<br />

of AS, moderate to severe plaque<br />

psoriasis and psoriatic arthritis.<br />

It targets interleukin-17A and<br />

may protect against bone loss<br />

by suppressing the function of<br />

osteoclasts and promoting the<br />

activity of osteoblasts.<br />

In the study, which will last two<br />

years, patients with the condition<br />

will undergo assessments, such<br />

as X-ray imaging of the skeleton to<br />

measure bone density and scoring<br />

of the spine for the presence<br />

of spurs or erosions. “What<br />

makes the disease so disabling<br />

is new bone formation that fuses<br />

the vertebrae,” says Susan M.<br />

Goodman, MD, Director of the<br />

Integrative <strong>Rheumatology</strong> and<br />

Orthopedic Center of Excellence<br />

and Attending Physician, who is<br />

leading the study. “Preliminary<br />

data supporting the use of the<br />

drug suggest that you don’t get<br />

“Preliminary data<br />

supporting the use<br />

of secukinumab, a<br />

monoclonal antibody<br />

used to treat<br />

autoimmune diseases,<br />

suggest that AS<br />

patients don’t get<br />

the bony proliferation<br />

as rapidly.”<br />

Susan M. Goodman, MD, Director<br />

of the Integrative <strong>Rheumatology</strong><br />

and Orthopedic Center of<br />

Excellence and Attending Physician<br />

the bony proliferation as rapidly.”<br />

Study participants will also<br />

be tested for markers of bone<br />

growth and resorption, as well as<br />

inflammation levels. If the results<br />

are positive, Dr. Goodman’s team<br />

will study the drug’s effects in a<br />

larger patient population.


Hospital for Special Surgery 11<br />

New possibilities for<br />

scleroderma treatment<br />

Scleroderma, one of the most<br />

life-threatening rheumatic<br />

diseases, is among the most<br />

challenging to understand and<br />

treat. But breakthrough research<br />

from the lab of Franck Barrat,<br />

PhD, Michael R. Bloomberg Chair<br />

and Senior Scientist, could lead<br />

to new therapeutic possibilities.<br />

Dr. Barrat’s research focuses<br />

on plasmacytoid dendritic<br />

cells (pDCs), which normally<br />

produce interferon to combat<br />

infection. In a study published in<br />

Science Translational Medicine<br />

in January <strong>2018</strong>, Dr. Barrat<br />

and <strong>HSS</strong> and international<br />

colleagues revealed that pDCs<br />

are chronically activated in<br />

scleroderma patients. They<br />

infiltrate the skin, where<br />

they trigger fibrosis and<br />

inflammation. “In the field<br />

of scleroderma, people have<br />

focused on how to stop<br />

fibrosis,” says Dr. Barrat.<br />

“But the correlation between<br />

inflammation and the main<br />

players that drive inflammation<br />

in fibrosis hasn’t been explored<br />

as much. This is pointing us in a<br />

direction that people had not<br />

looked at before.” As a result,<br />

the finding has generated<br />

considerable excitement from<br />

scientists and industry. “Some<br />

companies are looking really<br />

hard at this now,” says Dr. Barrat.<br />

Top: In our model, the secretion of CXCL4, a marker of systemic sclerosis<br />

(SSc), by patients’ plasmacytoid dendritic cells (pDCs) is due to the<br />

aberrant presence of TLR8 on these cells. CXCL4 potentiates the IFN<br />

production by pDCs, which promotes the autoimmune response and<br />

potentially fibrosis in patients. Depleting pDCs prevented disease in a<br />

preclinical model of scleroderma, suggesting that pDC is an essential<br />

cell type involved in the pathogenesis of SSc.<br />

Bottom, from left to right: Jessica K. Gordon, MD, Assistant Attending<br />

Physician, and Franck Barrat, PhD, Senior Scientist, as well as other<br />

<strong>HSS</strong> researchers, discovered that plasmacytoid dendritic cells (pDCs)<br />

are chronically activated in scleroderma patients, opening the door to<br />

investigations of new treatments.<br />

In a disease model, Dr. Barrat and<br />

colleagues showed that depleting<br />

pDCs prevented fibrosis. The<br />

research has helped to delineate<br />

important mechanisms of the<br />

aberrant cell activity. It has<br />

implicated a receptor on the<br />

surface of pDCs, called TLR8,<br />

as a culprit behind the cell<br />

activity. As a result, there may<br />

be several potential therapeutic<br />

targets and some existing drugs<br />

as possible candidates for<br />

study in scleroderma patients.<br />

“We have identified pDCs as<br />

potentially very important in the<br />

establishment and development<br />

of fibrosis,” says Dr. Barrat.<br />

“Now we can investigate new<br />

therapeutic options for targeting<br />

the cells, the ways they become<br />

activated, and some of the<br />

product they make.”<br />

These findings have led to new<br />

research funding, including<br />

nearly $1.8 million in grants from<br />

the National Institutes of Health<br />

and the Scleroderma Research<br />

Foundation. “What excites me<br />

is all of the possibilities that<br />

this research has opened up,”<br />

says Dr. Barrat. “We’ve put<br />

scleroderma on the map.”


12<br />

Division of <strong>Rheumatology</strong><br />

Using precision medicine<br />

to refine RA treatment<br />

The promise of precision<br />

medicine is to personalize<br />

therapy for patients with<br />

notoriously difficult-to-treat<br />

diseases such as rheumatoid<br />

arthritis (RA). To that end, <strong>HSS</strong><br />

scientists and physicians are<br />

investigating differences in RA<br />

patients’ responses to drugs as<br />

part of the five-year Accelerating<br />

Medicines Partnership (AMP)<br />

study, funded by the National<br />

Institutes of Health and<br />

partnering pharmaceutical<br />

companies. Our researchers<br />

are also investigating molecular<br />

pathways involved in flares.<br />

“We’re trying to discover new<br />

pathways, which could lead<br />

to new therapeutic targets,”<br />

says Lionel B. Ivashkiv, MD,<br />

Richard L. Menschel Chair and<br />

Chief Scientific Officer, David<br />

H. Koch Chair for Arthritis and<br />

Tissue Degeneration Research,<br />

and Director of the David Z.<br />

Rosensweig Genomics Research<br />

Center. “The ultimate goal is<br />

to use this information to help<br />

us choose the best drug for<br />

each patient at the beginning<br />

of treatment.”<br />

“Using advanced genomic<br />

technologies involving single<br />

cells, we are studying synovial<br />

tissue samples to find out how<br />

they relate to disease activity<br />

in the patient,” says Vivian P.<br />

Bykerk, MD, lead investigator<br />

of the RA study, Director of the<br />

Inflammatory Arthritis Center<br />

of Excellence and Associate<br />

Attending Physician. In the first<br />

phase of the study, which is<br />

now complete, our researchers<br />

were able to identify eight new<br />

cell types in RA joints using<br />

next-generation sequencing<br />

techniques. “These are<br />

subsets we’d want to target<br />

with new drugs,” says Laura<br />

Donlin, PhD, Co-Director of the<br />

Derfner Foundation Precision<br />

Medicine Laboratory and<br />

Assistant Scientist.<br />

In the second phase of the<br />

study, which is currently<br />

underway, our researchers are<br />

following a large cohort of RA<br />

patients who will have tissue<br />

samples taken before and after<br />

a new treatment is provided.<br />

Researchers will monitor them<br />

and look for commonalities<br />

that could serve as biomarkers<br />

for treatment response. “<strong>HSS</strong><br />

is a critical player in the AMP<br />

consortium because we provide<br />

unique scientific expertise<br />

and patient resources,” says<br />

Dr. Ivashkiv. “This study has<br />

changed the way we perform<br />

translational research, have<br />

scientific collaborations,<br />

and link clinical data with<br />

scientific data.”<br />

From left to right: Laura Donlin, PhD, Co-Director of the Derfner Foundation Precision Medicine Laboratory and<br />

Assistant Scientist, and Vivian P. Bykerk, MD, Director of the Inflammatory Arthritis Center of Excellence and<br />

Associate Attending Physician, review data for the AMP study.


Hospital for Special Surgery 13<br />

Baohong Zhao, PhD, Assistant Scientist, has identified two different methods for protecting against excessive<br />

osteoclast formation and bone destruction in disease models of osteoporosis and rheumatoid arthritis.<br />

Battling bone loss with<br />

a promising microRNAtargeting<br />

therapy<br />

Small non-coding RNA molecules<br />

that are approximately 21 to<br />

25 nucleotides in length, or<br />

microRNAs (miRNAs), regulate<br />

gene expression and functions<br />

in a variety of biological and<br />

pathological settings. While<br />

miRNA-based therapies have<br />

recently shown significant<br />

promise in the treatment of<br />

diseases like cancer, diabetes<br />

and hepatitis C, their use<br />

has been underexplored in<br />

osteoporosis and rheumatoid<br />

arthritis—until now. Baohong<br />

Zhao, PhD, Assistant Scientist<br />

in the Arthritis and Tissue<br />

Degeneration Program and The<br />

David Z. Rosensweig Genomics<br />

Research Center, has been using<br />

miRNA-sequencing techniques<br />

to profile genome-wide changes<br />

in miRNAs that are involved in<br />

the differentiation of osteoclasts.<br />

She has identified two different<br />

methods for protecting against<br />

excessive osteoclast formation<br />

and bone destruction in disease<br />

models of ovariectomy-induced<br />

osteoporosis and rheumatoid<br />

arthritis. One approach involves<br />

using a genetic knockout of<br />

miR-182 and the other involves<br />

a specific inhibitor targeting<br />

miR-182. “Our exciting genetic<br />

evidence and outstanding<br />

pharmacological results<br />

obtained from disease models<br />

provide a proof of concept for<br />

the efficacy of therapeutic<br />

targeting of miR-182 to prevent<br />

bone loss and highlight the<br />

translational implications<br />

of targeting miR-182 in<br />

treating osteolytic diseases,”<br />

says Dr. Zhao. “Now we are<br />

collaborating with translational<br />

researchers to see if we can<br />

develop more appropriate<br />

approaches to deliver a miR-<br />

182 inhibitor to patients with<br />

osteoporosis and arthritis to<br />

suppress bone loss.”<br />

While developing a safe and<br />

effective delivery system<br />

for this therapy is a priority,<br />

Dr. Zhao hopes that this<br />

miRNA will eventually be used<br />

as a biomarker to predict the<br />

progression of these diseases to<br />

prevent bone loss early on.


14<br />

Division of <strong>Rheumatology</strong><br />

Unraveling the molecular<br />

mysteries of lupus<br />

<strong>HSS</strong> Research Institute scientists<br />

are conducting studies at the<br />

molecular level to uncover new<br />

targets for lupus treatment.<br />

Alessandra Pernis, MD, The<br />

Peter Jay Sharp Chair in Lupus<br />

Research and Senior Scientist, is<br />

identifying molecular pathways<br />

of autoimmunity in the disease.<br />

Her work focuses on B cells,<br />

important players in autoimmune<br />

disease, including a subset called<br />

age-associated B cells, also<br />

known as ABCs. Dr. Pernis has<br />

discovered that ABC cells play two<br />

key roles in autoimmunity. “They<br />

can behave like B cells by making<br />

autoantibodies, and they can<br />

behave like inflammatory cells by<br />

making inflammatory cytokines,”<br />

she says. “Understanding the<br />

pathways that regulate these cells<br />

could represent a major advance<br />

in lupus as well as autoimmunity<br />

in general.”<br />

Using genome-wide transcriptional<br />

and epigenetic analysis of ABCs,<br />

Dr. Pernis and her team have<br />

uncovered the mechanism<br />

by which ABCs are regulated.<br />

“The majority of patients with<br />

autoimmunity are women, but<br />

until now we have not been able<br />

to find a molecular link,” says<br />

Dr. Pernis. “This is the first time<br />

we can see clear differences in<br />

males and females and how they<br />

behave at the transcriptional<br />

as well as the epigenetic level.”<br />

This research will enable us to<br />

uncover new treatment targets.<br />

Separately, Theresa T. Lu,<br />

MD, PhD, Associate Scientist,<br />

whose research is supported<br />

by the St. Giles Research Chair,<br />

is making headway toward<br />

understanding photosensitivity,<br />

whereby the sun’s ultraviolet<br />

(UV) light can trigger skin rashes<br />

as well as flares in patients.<br />

Photosensitivity affects 30 to<br />

60 percent of lupus patients.<br />

Dr. Lu’s research centers on<br />

Langerhans cells, immune<br />

cells in the top layer of skin. In<br />

a study published in Science<br />

Translational Medicine in <strong>2018</strong>,<br />

Dr. Lu, <strong>HSS</strong> colleagues and<br />

scientists at other institutions<br />

found that in healthy people,<br />

these cells protect the skin from<br />

UV damage. “With UV exposure,<br />

Langerhans cells normally<br />

activate ADAM17, a molecule<br />

that activates epidermal growth<br />

factor receptor (EGFR) ligands,<br />

helping skin cells to survive,”<br />

says Dr. Lu. In lupus models,<br />

Langerhans cells expressed less<br />

of the ADAM17 molecule, so<br />

they were less able to protect<br />

the skin. However, when EGFR<br />

ligand was added to the skin<br />

in lupus models, fewer lesions<br />

developed. More research is<br />

needed, but “stimulating EGFR<br />

in the skin may be an approach<br />

to ameliorating photosensitivity<br />

and systemic disease in lupus<br />

patients,” says Dr. Lu.<br />

Above: Langerhans cells (in red) sit<br />

among and protect the keratinocytes<br />

(in blue) in the skin epidermis.<br />

Photo Credit: William D. Shipman,<br />

PhD, Lu Lab, Autoimmunity and<br />

Inflammation Program<br />

Left, from left to right:<br />

Alessandra Pernis, MD, Senior<br />

Scientist, is identifying molecular<br />

pathways that play key roles in<br />

lupus, and Theresa T. Lu, MD,<br />

PhD, Associate Scientist, is<br />

gaining a better understanding of<br />

photosensitivity in lupus patients.


Hospital for Special Surgery 15<br />

From left to right: Jane E. Salmon, MD, Director of the Lupus and Antiphospholipid Syndrome Center of<br />

Excellence and Attending Physician, and Carl P. Blobel, MD, PhD, Director of the Arthritis and Tissue<br />

Degeneration Program and Senior Scientist, have discovered that iRhom2 may be an attractive new target for<br />

treating patients with lupus and hemophilic arthropathy.<br />

A promising new target for<br />

treating autoimmune diseases<br />

<strong>HSS</strong> scientists have identified a<br />

key protein that helps to regulate<br />

inflammation and could become<br />

a target for treating autoimmune<br />

diseases. The protein iRhom2<br />

appears to play a critical role<br />

in causing kidney damage in<br />

patients with lupus and seems to<br />

contribute to osteoporosis and<br />

joint damage in patients with<br />

hemophilic arthropathy.<br />

In a study published in the<br />

April <strong>2018</strong> issue of The Journal<br />

of Clinical Investigation, <strong>HSS</strong><br />

researchers and international<br />

colleagues found that<br />

inactivating iRhom2—which<br />

regulates ADAM17, whose<br />

substrates, such as TNF-α<br />

and heparin-binding EGF (HB-<br />

EGF), have been implicated in<br />

the pathogenesis of chronic<br />

kidney disease—prevented<br />

inflammation and irreversible<br />

scarring in lupus nephritis<br />

models. These same pathways<br />

were found to be upregulated<br />

in kidney biopsies from lupus<br />

patients. “The benefit of this<br />

approach is selective and<br />

simultaneous inhibition of two<br />

major pathological mediators<br />

of tissue damage in lupus,” says<br />

Jane E. Salmon, MD, Director of<br />

the Lupus and Antiphospholipid<br />

Syndrome Center of Excellence,<br />

Collette Kean Research Chair,<br />

and Attending Physician.<br />

In another study, published<br />

in Blood in September <strong>2018</strong>,<br />

<strong>HSS</strong> researchers found that<br />

inactivating iRhom2/TNF-α<br />

prevented the bone erosion and<br />

joint inflammation often seen in<br />

hemophilic arthropathy. “This is<br />

a major breakthrough because<br />

it points toward exciting new<br />

approaches for treatment of<br />

“The benefit of<br />

inactivating iRhom2<br />

is selective and<br />

simultaneous<br />

inhibition of two major<br />

pathological mediators<br />

of tissue damage in<br />

lupus.”<br />

Jane E. Salmon, MD, Director of<br />

the Lupus and Antiphospholipid<br />

Syndrome Center of Excellence<br />

and Attending Physician<br />

these two major manifestations<br />

of hemophilic arthropathy,” says<br />

Carl P. Blobel, MD, PhD, Director<br />

of the Arthritis and Tissue<br />

Degeneration Program, Virginia F.<br />

and William R. Salomon Chair<br />

in Musculoskeletal Research,<br />

and Senior Scientist.


16<br />

Division of <strong>Rheumatology</strong><br />

Investigating stem cells for<br />

repairing fibrotic skin<br />

“We think there<br />

are particular<br />

subpopulations of<br />

ADSCs that help to<br />

repair fibrotic skin, and<br />

we think there is a role<br />

for immune cells.”<br />

Theresa T. Lu, MD, PhD,<br />

Associate Scientist<br />

lymphotoxin to injected ADSCs<br />

would improve ADSC survival<br />

and effectiveness. They found<br />

that this combined regimen was<br />

able to partially reverse fibrosis.<br />

The question now is how exactly<br />

the ADSCs are working to repair<br />

fibrotic skin. “We think there<br />

are particular subpopulations<br />

of ADSCs that help to repair<br />

fibrotic skin, and we think there<br />

is a role for immune cells,”<br />

says Dr. Lu.<br />

Theresa T. Lu, MD, PhD, Associate Scientist, is studying adiposederived<br />

stromal cells (ADSCs) as a potential therapy for patients with<br />

scleroderma and other diseases.<br />

One of the major challenges<br />

in managing scleroderma<br />

patients is the lack of effective<br />

treatments for skin fibrosis.<br />

This condition is not only<br />

painful and can lead to severe<br />

joint contractures and loss<br />

of hand function, but it can<br />

also cause asymmetric limb<br />

or facial growth in children.<br />

In addition, wound healing is<br />

compromised. Theresa T. Lu,<br />

MD, PhD, Associate Scientist<br />

and a member of the Division<br />

of Pediatric <strong>Rheumatology</strong>,<br />

and members of her lab<br />

have been studying adiposederived<br />

stromal cells (ADSCs),<br />

which contribute to the repair<br />

and regeneration of tissues,<br />

including skin. Her lab has found<br />

that ADSC numbers are reduced<br />

in skin fibrosis—most likely<br />

due to cell death—suggesting<br />

that effective replenishment of<br />

these cells could repair fibrotic<br />

skin and be a possible therapy<br />

in a multipronged approach for<br />

scleroderma patients. In a study<br />

published in The Journal<br />

of Clinical Investigation,<br />

Dr. Lu and colleagues found that<br />

certain immune cells known<br />

as dendritic cells help ADSCs<br />

survive in fibrotic skin. There is<br />

an interest in the scleroderma<br />

field in injecting ADSCs to<br />

repair the skin, but one of the<br />

limitations is that ADSCs do<br />

not survive a long time. Dr. Lu’s<br />

group reasoned that adding<br />

the dendritic cell-derived signal<br />

In <strong>2017</strong>, Dr. Lu received a<br />

grant from the Scleroderma<br />

Foundation to determine which<br />

ADSC subpopulations are<br />

reparative and which ones may<br />

be harmful. Dr. Lu also recently<br />

received funding from the nonprofit<br />

organization A Lasting<br />

Mark to begin to examine the<br />

role of immune cells in mediating<br />

ADSC effects. One future goal<br />

is to find out whether ADSC<br />

populations in children’s skin<br />

are more or less susceptible to<br />

damage by fibrosis compared<br />

to adults’ skin. “We know that<br />

children’s skin heals better<br />

than adults’ skin,” says Dr. Lu.<br />

“Determining whether there<br />

are ADSC subpopulations that<br />

are more protective in children<br />

will help us gain a better<br />

understanding of scleroderma—<br />

and potentially improve therapies<br />

for patients.”


Hospital for Special Surgery 17<br />

Optimizing postsurgical outcomes<br />

Ensuring that patients are<br />

medically ready for surgery can<br />

help improve outcomes—and<br />

their overall experience at <strong>HSS</strong>.<br />

“Our orthopaedic surgeons were<br />

one of the first groups in the<br />

nation to embrace the concept<br />

of co-management of the<br />

patient,” says Linda A. Russell,<br />

MD, Director of the Division of<br />

Perioperative Medicine, Anne<br />

and Joel Ehrenkranz Chair in<br />

Perioperative Medicine, and<br />

Associate Attending Physician.<br />

“Every single patient who’s<br />

admitted to <strong>HSS</strong> has a surgeon<br />

and a medical physician familiar<br />

with the perioperative care of the<br />

orthopaedic patient.”<br />

Physicians in perioperative<br />

medicine, a field that has<br />

emerged over the last 15 years,<br />

are assigned to follow specific<br />

patients to help ensure continuity<br />

of care. “Perioperative medicine<br />

physicians see patients before<br />

surgery and try to get them in<br />

as good a shape as they can,”<br />

says Dr. Russell. “They also see<br />

patients when they’re in the<br />

hospital. Then they’re available to<br />

patients for up to 90 days or so<br />

after the surgery for any related<br />

medical conditions.”<br />

Prior to surgery, <strong>HSS</strong> patients<br />

are carefully assessed to identify<br />

and address any medical<br />

problems that could impact<br />

surgical outcomes, including<br />

uncontrolled diabetes, cardiac<br />

issues, skin infections and, most<br />

recently, obesity. “There’s a lot<br />

of research that says if your<br />

BMI is 40 or higher, your risk of<br />

postoperative complications is<br />

much higher,” says Dr. Russell.<br />

In July <strong>2018</strong>, the Division of<br />

Perioperative Medicine added<br />

a weight-loss program to help<br />

obese patients lose at least<br />

5 percent of their total body<br />

weight before surgery. “We are<br />

probably the first hospital in the<br />

country that’s really focused on<br />

perioperative weight loss,” says<br />

Dr. Russell. Obese patients see a<br />

weight-management physician<br />

and nutritionist at <strong>HSS</strong> at least<br />

two to three months before<br />

surgery to jump-start healthy<br />

eating habits and weight loss.<br />

“We continue to follow up with<br />

these patients after surgery<br />

as well for long-term weight<br />

management,” says Caroline A.<br />

Andrew, MD, Assistant Attending<br />

Physician, who directs the<br />

weight management program<br />

at <strong>HSS</strong>. “Losing weight after<br />

surgery helps with recovery<br />

and can reduce postsurgical<br />

osteoarthritis, in addition to<br />

improving other comorbidities<br />

of obesity.”<br />

Many obese patients have<br />

diabetes, so perioperative<br />

medicine practitioners help them<br />

get the condition under control.<br />

These patients may work with<br />

an endocrinologist to achieve<br />

the healthiest A1C levels prior<br />

to surgery.<br />

Dr. Russell’s team of physicians,<br />

physician assistants and nurse<br />

practitioners also work to<br />

optimize the use of opioids and<br />

other pain medications before<br />

and after surgery. “A chronic<br />

pain physician sees patients who<br />

take opioids daily to taper pain<br />

medications preoperatively and<br />

design the postoperative pain<br />

program,” she says.<br />

After surgery, wound issues,<br />

leg swelling and other<br />

complications can be treated<br />

by the perioperative medicine<br />

staff, saving the patient a trip<br />

to an urgent care facility or the<br />

emergency room.<br />

From left to right: Caroline A. Andrew, MD, Assistant Attending Physician,<br />

and Linda A. Russell, MD, Director of the Division of Perioperative<br />

Medicine and Associate Attending Physician, collaborate to ensure<br />

patients are medically ready for surgery.<br />

Over the last decade, the<br />

Division of Perioperative<br />

Medicine has developed<br />

numerous surgical guidelines,<br />

including rules about when<br />

medications should be stopped<br />

preoperatively and started<br />

postoperatively; which patients<br />

with diabetes can be cleared for<br />

the OR; and which patients are<br />

candidates for bilateral knee<br />

replacements. “The goal is to<br />

help decrease postoperative<br />

complications and readmission<br />

rates,” says Dr. Russell. “We also<br />

hope the guidelines can provide<br />

a better patient experience.”


18<br />

Division of <strong>Rheumatology</strong><br />

Complex Cases<br />

Case 1<br />

Eosinophilic Fasciitis<br />

Associated with Cancer<br />

Immunotherapy<br />

19<br />

Case 2<br />

Inflammatory Erosive<br />

Arthritis and Tendon<br />

Rupture in Sarcoidosis<br />

20<br />

Case 3<br />

Severe Lupus with<br />

Nephritis and Catatonia;<br />

Excellent Response to<br />

Aggressive Treatment<br />

21<br />

Case 4<br />

Subacute Bacterial<br />

Endocarditis Masquerading<br />

as Polymyalgia Rheumatica<br />

22<br />

Case 5<br />

Retiform Purpura<br />

Initially Concerning for<br />

Granulomatosis with<br />

Polyangiitis<br />

23<br />

<strong>HSS</strong> rheumatologists frequently receive calls from physicians around the world<br />

asking for advice in treating complex cases. Recent calls have come from Sri Lanka,<br />

France, Kuwait and Puerto Rico. The following five cases are excerpted from Grand<br />

Rounds from <strong>HSS</strong>: Management of Complex Cases, which includes follow-up data<br />

and a clinical treatment discussion.<br />

Visit the entire list of Case references online at hss.edu/complexcases


Hospital for Special Surgery 19<br />

Case 1 presented by David R. Fernandez, MD, PhD, and Anne R. Bass, MD<br />

Eosinophilic Fasciitis Associated<br />

with Cancer Immunotherapy<br />

Case <strong>Report</strong><br />

A 48-year-old man was referred for<br />

complaints of “stiffness” in his legs. One<br />

year prior to presentation he was diagnosed<br />

with stage IV lung adenocarcinoma (EGFR<br />

exon 19 deletion) with lung metastases. He<br />

was treated with nivolumab and erlotinib and<br />

experienced 50 percent tumor regression.<br />

Six months into treatment, he began to<br />

note tightness and pain in his thigh and calf<br />

muscles, shoulders, ankles and wrists. He<br />

had trouble walking and rising from a chair.<br />

He also noted calf cramps and leg swelling.<br />

He denied Raynaud’s phenomenon.<br />

Laboratory testing revealed CPK 933<br />

and an absolute eosinophil count of 700.<br />

CPK normalized without intervention,<br />

but total eosinophils climbed to 3,500<br />

over the next three months. Nivolumab<br />

was discontinued and the patient was<br />

referred to rheumatology. Examination<br />

revealed thickening of the fascia of the<br />

forearms and of the legs below the knees,<br />

which limited mobility at the elbows,<br />

wrists, knees and ankles. The patient<br />

could barely pronate or supinate the<br />

right elbow. Extension of the right wrist<br />

was limited to 20° due to tightening<br />

of the fascia, and there were flexion<br />

contractures of both knees. A “groove<br />

sign” was noted over the left leg and right<br />

forearm (Figure 1). The superficial skin,<br />

nailfold capillaries and joints themselves<br />

were normal. The patient was treated<br />

with high-dose oral corticosteroids with<br />

moderate improvement. Deep skin biopsy<br />

was consistent with eosinophilic fasciitis<br />

(Figure 2). The patient continued treatment<br />

with high-dose corticosteroids and<br />

experienced gradual softening of the fascia<br />

and increased mobility of the arms and,<br />

to a lesser degree, the legs. Methotrexate<br />

was added to his regimen to enable a rapid<br />

steroid taper prior to lobectomy for residual<br />

disease. The patient continued to require<br />

low doses of prednisone 10 months after the<br />

onset of his disease.<br />

Discussion<br />

Eosinophilic fasciitis is a rare disease of<br />

unknown etiology. It is characterized by<br />

peripheral eosinophilia and induration of<br />

the deep fascia underlying the skin. [1] It is<br />

generally steroid-responsive, but it often<br />

requires prolonged therapy and not all<br />

changes may be reversible. Eosinophilic<br />

fasciitis can be associated with trauma,<br />

drugs, infections, autoimmune conditions<br />

and cancer, but to our knowledge, this<br />

is the first case associated with cancer<br />

immunotherapy. [2] The timing of this<br />

patient’s presentation suggests that the<br />

Figure 1: Photograph of the left leg demonstrating a “groove sign” along the path of a superficial vein.<br />

nivolumab and not the cancer itself<br />

was responsible.<br />

Nivolumab is one of a new class of<br />

medications directed at enhancing<br />

antitumor immunity by inhibiting normal<br />

checkpoints on immune cell function.<br />

Immune checkpoint blockade has<br />

revolutionized the treatment of many<br />

cancers but at the expense of frequent<br />

immune-mediated side effects. [3] The<br />

programmed cell death-1 (PD-1) receptor<br />

is an immune checkpoint molecule<br />

expressed on activated T cells, which<br />

transmits inhibitory signals to T cells and<br />

enhances the generation of regulatory<br />

T cells. High expression of PD-1 on T<br />

cells can be seen in the setting of chronic<br />

infections and is associated with a<br />

phenomenon called “exhaustion,” which<br />

is associated with poor T cell function and<br />

high expression of inhibitory molecules<br />

on T cells. [4]<br />

Normally, cells in the peripheral organs can<br />

increase expression of programmed cell<br />

death ligand 1 (PD-L1) in certain settings.<br />

However, cancer cells also frequently<br />

express PD-L1, leading to diminished<br />

T cell-directed immune responses<br />

against tumors. Nivolumab blocks PD-1<br />

signaling, leading to enhanced antitumor<br />

immunity, but has important side effects<br />

related to induction of autoimmunity,<br />

such as rash, hepatitis, pneumonitis and<br />

thyroiditis. Interestingly, the autoimmune<br />

manifestations associated with<br />

immunotherapy can be short-lived when<br />

treated with steroids or tumor necrosis<br />

factor inhibitors. They generally lack<br />

the characteristic autoantibodies<br />

associated with spontaneous-onset<br />

rheumatologic diseases. [5]<br />

This case implies a potential role for PD-1<br />

signaling as a key tolerance mechanism,<br />

Figure 2: Deep skin biopsy demonstrating<br />

broad, hyalinized collagen bundles within the<br />

fascia with deposition of mucin and a<br />

lymphocytic and plasma cell infiltrate.<br />

preventing the development of eosinophilic<br />

fasciitis in susceptible individuals.<br />

References<br />

1. Lebeaux D, Frances C, Barete S, et al.<br />

Eosinophilic fasciitis (Shulman disease): new<br />

insights into the therapeutic management<br />

from a series of 34 patients. <strong>Rheumatology</strong><br />

2012;51(3):557–61.<br />

2. Pinal-Fernandez I, Selva-O’Callaghan A., Grau<br />

JM. Diagnosis and classification of eosinophilic<br />

fasciitis. Autoimmun Rev 2014;13(4–5):379–82.<br />

3. Haanen JB a. G, Thienen H Van, Blank CU.<br />

Toxicity Patterns With Immunomodulating<br />

Antibodies and Their Combinations. Semin<br />

Oncol 2015;42(3):423–8.<br />

4. Wherry EJ, Kurachi M. Molecular and cellular<br />

insights into T cell exhaustion. Nat Rev<br />

Immunol 2015;15(8):486–99.<br />

5. Villadolid J, Amin A. Immune checkpoint<br />

inhibitors in clinical practice: update on<br />

management of immune-related toxicities.<br />

Transl lung cancer Res 2015;4(5):560–75.<br />

Author Disclosures<br />

Dr. David R. Fernandez does not have a financial<br />

interest or relationship with the manufacturers<br />

of products or services.<br />

Dr. Anne R. Bass does not have a financial<br />

interest or relationship with the manufacturers<br />

of products or services.


20<br />

Division of <strong>Rheumatology</strong><br />

Case 2 presented by Arthur M. F. Yee, MD, PhD<br />

Inflammatory Erosive Arthritis<br />

and Tendon Rupture in Sarcoidosis<br />

Case <strong>Report</strong><br />

A 64-year-old woman was referred to<br />

the <strong>HSS</strong> Division of <strong>Rheumatology</strong> for a<br />

second opinion regarding chronic pain<br />

and swelling of the right wrist and acute<br />

loss of extension of the right small finger.<br />

The patient had a 20-year history of<br />

sarcoidosis, initially presenting as right<br />

eye uveitis, retinal vasculitis and acute<br />

polyarthritis. Evaluation at the time revealed<br />

mediastinal lymphadenopathy, the biopsy<br />

of which led to the diagnosis of sarcoidosis.<br />

The patient was treated chronically for<br />

many years with varying dosages of<br />

prednisone before the eventual addition of<br />

weekly methotrexate enabled subsequent<br />

withdrawal of all corticosteroid therapies.<br />

After several months of clinical remission,<br />

methotrexate was also discontinued. She<br />

remained symptom-free for several months<br />

before developing insidiously progressive<br />

chronic right wrist pain and swelling. MRI<br />

demonstrated extensive inflammatory<br />

arthritis and tenosynovitis with marked<br />

erosions in the distal radius and ulna and<br />

encroachment upon the extensor tendons<br />

(Figure 1). She was treated with nonsteroidal<br />

anti-inflammatory drugs and periodic<br />

short courses of systemic corticosteroids<br />

with marginal responses. After two years<br />

of waxing and waning inflammatory right<br />

wrist arthritis, she developed an acutely<br />

dropped right small finger, prompting<br />

referral to <strong>HSS</strong>.<br />

Physical examination was notable for<br />

moderate swelling, tenderness and<br />

warmth of the right wrist; loss of extension<br />

of the right small finger; and baseline loss<br />

of visual acuity of the right eye without<br />

active inflammation. There was no<br />

evidence of active synovitis elsewhere.<br />

Complete blood count, metabolic panel,<br />

erythrocyte sedimentation rate, C-reactive<br />

protein, angiotensin-converting enzyme,<br />

rheumatoid factor, and anti-cyclic<br />

citrullinated protein antibodies were normal<br />

or negative. Large erosions of the distal<br />

right ulna and radius were evident on plain<br />

radiography (Figure 2).<br />

An immediate consultation to the Hand<br />

and Upper Extremity Service was made,<br />

and the patient underwent prompt<br />

synovectomy and repair of the ruptured<br />

extensor tendons. Histopathology<br />

showed inflammatory synovitis and<br />

excluded infectious etiologies. After<br />

a brief, uncomplicated postoperative<br />

period, oral methotrexate was initiated<br />

and was expeditiously titrated to 15 mg<br />

weekly. The patient has done well for five<br />

years without recurrence of inflammatory<br />

arthritis or uveitis.<br />

Discussion<br />

Sarcoidosis is an enigmatic systemic<br />

inflammatory disorder characterized<br />

by non-caseating granulomatous<br />

inflammation of potentially any tissue.<br />

While nonspecific arthralgias are<br />

common, frank inflammatory arthritis<br />

is infrequent and can be categorized<br />

as acute or chronic. In one recent<br />

long-term North American series,<br />

one‐tenth of all patients developed acute<br />

inflammatory arthritis/periarthritis, [1]<br />

typically oligoarticular or polyarticular,<br />

although prevalence may range widely<br />

depending on the population examined.<br />

The most stereotypical presentation<br />

is Lofgren’s syndrome (acute bilateral<br />

ankle periarthritis, erythema nodosum,<br />

and uveitis), which can be quite painful<br />

but is fortunately self-limited and<br />

nondestructive. [2] In most cases, acute<br />

sarcoid arthritis is reminiscent of<br />

exacerbations of inflammatory arthritis<br />

seen in other systemic conditions,<br />

likely reflecting nonspecific systemic<br />

inflammatory processes rather than<br />

direct granulomatous infiltration in the<br />

joint. In contrast, infiltrative chronic<br />

granulomatous synovitis, as seen<br />

in the presented case, is much less<br />

common, is sometimes quite resistant<br />

to medical therapy, and occasionally<br />

leads to local tissue injury. [3] A diverse<br />

array of clinical scenarios, such as<br />

bursitis, enthesopathies, sacroiliitis, and<br />

carpal tunnel syndrome, resulting from<br />

granulomatous inflammation have<br />

been reported. [4]<br />

The challenges facing <strong>HSS</strong> physicians in<br />

this case were to first repair the ruptured<br />

fifth extensor mechanism and then to<br />

implement a medical regimen to prevent<br />

recurrence. After adequate postoperative<br />

healing and rehabilitation, medical<br />

management was initiated.<br />

Steroid-sparing agents are increasingly<br />

being used for the treatment of sarcoidosis,<br />

but none are formally approved. Nonetheless,<br />

methotrexate has emerged as the preferred<br />

steroid-sparing agent. [5] Controlled clinical<br />

studies of methotrexate in the treatment<br />

of sarcoidosis have demonstrated efficacy<br />

in specific common phenotypes, such as<br />

pulmonary disease, cardiomyopathy and<br />

panuveitis. There are ample case reports<br />

and series of use in other manifestations,<br />

such as involvement of the skin, bone,<br />

neuraxis, muscle, etc. [4]<br />

The acknowledged efficacy of methotrexate<br />

has led to the development of proposed<br />

guidelines for its use in sarcoidosis. [5] Many<br />

recommendations echo those for the use<br />

of methotrexate in rheumatoid arthritis, but<br />

some reflect the personal practice and expert<br />

opinion of “sarcoidologists.” It is very notable<br />

that the majority of experts recommended<br />

weekly methotrexate maintenance dosages<br />

of no more than 15 mg. Therefore, it is<br />

possible that methotrexate is underutilized<br />

in sarcoidosis.<br />

References<br />

1. Ungprasert P, Crowson C S, et al. Clinical<br />

characteristics of sarcoid arthropathy: a<br />

population-based study. Arthritis Care Res<br />

2016; 68: 695–9.<br />

2. Mana J, Gomez-Vaquero C, et al. Lofgren’s<br />

syndrome revisited: a study of 186 patients.<br />

Am J Med 1999; 107: 240–5.<br />

3. Chu A, Ginat D, et al. Chronic sarcoid arthritis<br />

presenting as an intra-articular knee mass.<br />

J Clin Rheumatol 2009; 15: 190–2.<br />

4. Yee AMF. Sarcoidosis: rheumatology<br />

perspective. Best Pract Res Clin Rheumatol.<br />

5. Cremers JP, Drent M, et al. Multinational<br />

evidence-based World Association of<br />

Sarcoidosis and Other Granulomatous<br />

Disorders recommendations for the use<br />

of methotrexate in sarcoidosis: integrating<br />

systematic literature research and expert<br />

opinion of sarcoidologists worldwide. Current<br />

Opin Pulm Med 2013; 19: 545–61.<br />

Author Disclosures<br />

Dr. Yee is on a Speaker’s Bureau and has<br />

participated on Advisory Boards for Bristol-<br />

Myers Squibb and has stock shares in Abbvie<br />

and Pfizer.<br />

Figure 1: Fat-suppressed MRI (coronal view) of<br />

the dorsum of the right wrist/hand. Bulky<br />

inflammatory synovitis (enclosed in red oval)<br />

impinges on the extensor digitorum longus<br />

tendon (arrows) to the small finger.<br />

Figure 2: Frontal view of the right wrist/hand.<br />

Large erosions (encircled) are evident in the distal<br />

radius and ulna.


Hospital for Special Surgery 21<br />

Case 3 presented by Naveed Chaudhry, MD, Steven P. Salvatore, MD,<br />

and Kyriakos A. Kirou, MD<br />

Severe Lupus with Nephritis and<br />

Catatonia; Excellent Response to<br />

Aggressive Treatment<br />

Case <strong>Report</strong><br />

A 34-year-old woman was diagnosed with<br />

systemic lupus erythematosus (SLE) in<br />

2011 after she presented with arthritis,<br />

photosensitivity, alopecia and oral ulcers.<br />

She was treated with short courses of<br />

prednisone. Her serologic profile was<br />

positive for ANA, anti-dsDNA, anti-Sm<br />

and anti-RNP antibodies. In January 2013,<br />

she was admitted to another hospital for<br />

left popliteal vein deep vein thrombosis<br />

(DVT) and pulmonary embolism (PE). She<br />

was also noted to have nephrotic range<br />

proteinuria and pleuropericarditis. She was<br />

treated with anticoagulation (enoxaparin to<br />

warfarin) and prednisone 60 mg/day with<br />

improved symptoms.<br />

She first presented to us in February 2013<br />

with marked leg edema and dyspnea on<br />

exertion. Past medical and family histories<br />

were unremarkable. Her blood pressure<br />

was 112/82, pulse 85, and she had 2+ leg<br />

edema. Her Hgb was 13.9 gm/dl, serum<br />

creatinine (Cr) 0.64 mg/dL, albumin<br />

1.7 gm/dl, cholesterol 366 mg/dl, ESR 72<br />

mm/hr, C3 167 mg/dL, C4 64 mg/dL, and<br />

anti-DNA 2+.<br />

Her urine sediment was bland, but her urine<br />

protein-creatinine ratio (UPCR) was 5.2.<br />

She was on prednisone 50 mg/day, and<br />

mycophenolate mofetil (MMF) was added to<br />

her regimen.<br />

Her edema got much worse, however,<br />

and she was readmitted three weeks later<br />

(March 2013) with severe anasarca. She<br />

received IV methylprednisolone 500 mg<br />

daily for three days and, after switching<br />

from warfarin to heparin, a renal biopsy<br />

was performed. This revealed ISN/RPS<br />

Class V membranous lupus nephritis<br />

(Figures 1–2). MMF was discontinued<br />

because of diarrhea, and she was treated<br />

with IV cyclophosphamide (IVCY). The<br />

patient was discharged on dexamethasone<br />

9 mg, losartan 25 mg, bumetanide 1 mg,<br />

warfarin, atorvastatin 40 mg, calcium,<br />

vitamin D3, and alendronate 70 mg weekly.<br />

She was readmitted to the hospital in early<br />

April 2013 after she developed symptoms<br />

of mania, depression and psychosis with<br />

paranoia. She was also noted to have<br />

severe edema and pneumonia.<br />

The patient was treated with IV antibiotics<br />

and diuretics, and glucocorticoids<br />

were continued. After treatment with<br />

haloperidol, she developed catatonia<br />

with motor rigidity, immobility, staring,<br />

mutism and negativism. She was not<br />

hyperthermic, and creatine kinase and<br />

electroencephalogram were normal.<br />

She was treated with IV lorazepam 2 mg,<br />

and high-dose glucocorticoids were<br />

continued. Her mental status normalized<br />

after a few days. Pulmonary infiltrates<br />

cleared 11 days later and she was given<br />

her second IVCY dose as well as IV<br />

rituximab 1,000 mg. She was discharged<br />

on prednisone 40 mg daily with a slow<br />

taper. She continued with monthly IVCY<br />

for a total of five cycles (the last one<br />

was in August 2013) and had gradual<br />

improvement of proteinuria and edema.<br />

By September 2013, her UPCR was<br />

0.37 and her albumin had normalized.<br />

She was started on azathioprine<br />

50 mg BID as maintenance treatment<br />

while the prednisone was tapered to<br />

5 mg/day. Warfarin was discontinued<br />

without recurrent thrombosis. Her<br />

antiphospholipid antibodies, including<br />

lupus anticoagulant, have remained<br />

negative. The patient was able to go back<br />

to her job full-time.<br />

Discussion<br />

We have described a complex case of<br />

lupus with active disease in several<br />

organs and excellent response to prompt<br />

and aggressive management. The<br />

patient first presented with nephrosis<br />

and secondary DVT-PE. Then she<br />

developed pleuropericarditis and, finally,<br />

neuropsychiatric (NP) SLE with bipolar<br />

symptoms, psychosis and catatonia.<br />

Although glucocorticoids may cause<br />

psychosis, it was unlikely they were the<br />

culprit here, as they had been tolerated<br />

well. The patient’s NP symptoms occurred<br />

concurrently with disease activity in<br />

other organs. Catatonia is a rare NP<br />

manifestation of SLE, often associated<br />

with an underlying bipolar disorder. [1]<br />

It may be triggered by antipsychotics<br />

like haloperidol and often responds to<br />

lorazepam, as in our case. Treatment of<br />

lupus with glucocorticoids, IVCY and/or<br />

plasma exchange is also effective. [1]<br />

Pure membranous lupus nephritis,<br />

when accompanied by nephrotic range<br />

proteinuria, is also a severe manifestation<br />

of SLE and demands treatment with<br />

MMF, IVCY or cyclosporine. [2, 3] We added<br />

rituximab, as it is often effective in severe<br />

lupus as well as NPSLE. [4]<br />

References<br />

1. Grover S, Parakh P, Sharma A et al. Catatonia<br />

in systemic lupus erythematosus: a case<br />

report and review of literature. Lupus 2013;<br />

22: 634–8.<br />

2. Austin HA, 3rd, Illei GG, Braun MJ, Balow JE.<br />

Randomized, controlled trial of prednisone,<br />

cyclophosphamide, and cyclosporine in<br />

lupus membranous nephropathy. J Am Soc<br />

Nephrol 2009; 20: 901–11.<br />

3. Radhakrishnan J, Moutzouris DA, Ginzler EM<br />

et al. Mycophenolate mofetil and intravenous<br />

cyclophosphamide are similar as induction<br />

therapy for class V lupus nephritis. Kidney Int<br />

2010; 77: 152–60.<br />

4. Tokunaga M, Saito K, Kawabata D et al.<br />

Efficacy of rituximab (anti-CD20) for<br />

refractory systemic lupus erythematosus<br />

involving the central nervous system. Ann<br />

Rheum Dis 2007; 66: 470–5.<br />

Author Disclosures<br />

Dr. Naveed Chaudhry does not have a financial<br />

interest or relationship with the manufacturers<br />

of products or services.<br />

Dr. Kyriakos A. Kirou does not have a financial<br />

interest or relationship with the manufacturers<br />

of products or services.<br />

Dr. Steven P. Salvatore does not have a financial<br />

interest or relationship with the manufacturers<br />

of products or services.<br />

Figure 1: C3 staining by immunofluorescence<br />

microscopy showing global 3+ capillary wall<br />

deposits (40x).<br />

Figure 2: Electron microscopy revealing global<br />

subepithelial electron dense deposits (arrows).


22<br />

Division of <strong>Rheumatology</strong><br />

Case 4 presented by Alana B. Levine, MD<br />

Subacute Bacterial<br />

Endocarditis Masquerading<br />

as Polymyalgia Rheumatica<br />

Case <strong>Report</strong><br />

A 70-year-old woman was referred to the<br />

<strong>HSS</strong> Division of <strong>Rheumatology</strong> for a second<br />

opinion regarding polymyalgia rheumatica<br />

(PMR). She had been diagnosed with PMR<br />

at another hospital eight months prior<br />

to presentation based on fatigue and an<br />

elevated ESR. Her fatigue resolved rapidly<br />

after starting prednisone 20 mg daily,<br />

which was tapered off over the course<br />

of two months. Fatigue recurred after<br />

discontinuing steroids and the patient was<br />

restarted on prednisone and, eventually,<br />

methotrexate.<br />

One month prior to presentation at <strong>HSS</strong>,<br />

the patient developed a new non-painful,<br />

non-pruritic leg rash. Skin biopsy revealed<br />

leukocytoclastic vasculitis. At the time, she<br />

was taking prednisone 5 mg daily, which<br />

was increased to 40 mg daily, and the<br />

rash resolved.<br />

On presentation to <strong>HSS</strong>, the patient<br />

complained of severe fatigue, nocturnal<br />

fevers to 101°F, night sweats and anorexia<br />

with a 20-pound weight loss.<br />

The patient’s past medical history was<br />

significant for mitral valve prolapse and<br />

osteopenia. She had dental work one<br />

month prior to the PMR diagnosis and had<br />

two caesarian sections in the past. Her<br />

medications included methylprednisolone<br />

4 mg daily, rabeprazole 20 mg daily, and<br />

calcium supplementation.<br />

On physical examination, the patient was in<br />

no acute distress. Her blood pressure was<br />

110/65 mm Hg, pulse 112, and temperature<br />

100.2°F. She weighed 104 pounds and was<br />

62 inches tall. Her exam was remarkable for<br />

a loud systolic murmur, heard throughout<br />

the precordium but most pronounced at<br />

the apex. She had fingernail clubbing and<br />

several non-blanching 2-mm palpable<br />

purpuric lesions over the shins. Her lung<br />

exam was unremarkable, pulses were<br />

strong throughout and without bruits,<br />

and she had no synovitis. The differential<br />

diagnosis at that time included systemic<br />

vasculitis, malignancy and endocarditis.<br />

Labs were significant for hemoglobin<br />

8.7 g/dL, serum creatinine 2.2 mg/dL<br />

(0.9 mg/dL one month prior), rheumatoid<br />

factor 222 IU/mL, ESR 53 mm/hr, CRP<br />

125 mg/L, C3 31 mg/dL, and C4 7 mg/dL.<br />

Urinalysis showed 2+ protein, 3+ blood,<br />

>30 WBC/hpf and >30 RBC/hpf.<br />

Antinuclear antibodies, double-stranded<br />

DNA antibodies, extractable nuclear<br />

antigens, and anti-CCP antibodies<br />

were negative.<br />

The patient was admitted for evaluation of<br />

acute renal failure. Multiple sets of blood<br />

cultures were drawn and held for slowgrowing<br />

organisms, and the patient was<br />

started on broad-spectrum antibiotics.<br />

A renal ultrasound was unremarkable. A<br />

transthoracic echocardiogram showed<br />

severe mitral regurgitation with a<br />

1.9 x 1.8 cm vegetation on the posterior<br />

mitral leaflet. Blood cultures grew<br />

gram-variable rods, which were later<br />

speciated as Suttonella indologenes.<br />

Due to the size of the vegetation and<br />

severe mitral regurgitation, the patient<br />

was recommended for mitral valve<br />

replacement surgery.<br />

Discussion<br />

Subacute bacterial endocarditis may cause<br />

a constellation of symptoms, including<br />

fatigue and malaise, fevers, valvular<br />

dysfunction, renal failure secondary to<br />

glomerulonephritis, and leukocytoclastic<br />

vasculitis. Suttonella indologenes is a<br />

gram-negative, rod-shaped bacterium<br />

found in normal respiratory tract flora, but it<br />

[1, 2]<br />

has been known to cause endocarditis.<br />

References<br />

1. Ozcan F, Yildiz A, Ozlu MF, et al. A case of fatal<br />

endocarditis due to Suttonella indologenes.<br />

Anadolu Kardiyol Derg 2011;11:85–7.<br />

2. Yang EH, Poon K, Pillutla P, Budoff MJ, Chung<br />

J. Pulmonary embolus caused by Suttonella<br />

indologenes prosthetic endocarditis<br />

in a pulmonary homograft. J Am Soc<br />

Echocardiogr 2011;24:592 e1–3.<br />

Author Disclosures<br />

Dr. Alana B. Levine does not have a financial<br />

interest or relationship with the manufacturers<br />

of products or services.<br />

Figure 1: Transthoracic echocardiogram showing<br />

1.9 x 1.8 cm vegetation on the mitral valve.<br />

Figure 2: Transesophageal echocardiogram<br />

showing vegetation and flail posterior<br />

mitral leaflet.<br />

Figure 3: Transesophageal echocardiogram<br />

showing severe mitral regurgitation and<br />

perforation of the posterior mitral leaflet.


Hospital for Special Surgery 23<br />

Case 5 presented by Danielle Ramsden-Stein, MD, Sonali Narain MD, MPH, and<br />

Michael D. Lockshin, MD<br />

Retiform Purpura Initially<br />

Concerning for Granulomatosis<br />

with Polyangiitis<br />

Case <strong>Report</strong><br />

A 47-year-old woman with recurrent<br />

sinusitis and bronchitis, intermittent febrile<br />

rashes and depression presented to an<br />

outside hospital with rapidly progressing<br />

skin rash after eating at a seafood<br />

restaurant. She had bruises on her left<br />

shoulder, left thigh and under her left<br />

breast accompanied by fever to 102°F,<br />

arthralgias and fatigue. The rash worsened<br />

within 24 hours to involve her upper and<br />

lower extremities, cheeks, ears, breasts<br />

and abdomen. She denied trauma, toxic<br />

exposures, new medications, sick contacts,<br />

or recent travel.<br />

On examination, she had saddle nose<br />

deformity and necrotizing purpura<br />

with bullae developing over the<br />

dependent areas. She had proteinuria<br />

and pancytopenia. Lumbar puncture<br />

was negative, but chest X-ray showed<br />

left lower lobe consolidation. She was<br />

started on ceftriaxone/vancomycin. She<br />

developed a cough with blood-tinged<br />

sputum and gross hematuria. Head CT<br />

scan showed pansinusitis. Intravenous<br />

methylprednisolone 40 mg every six hours<br />

was prescribed. The lesions progressed<br />

from pruritic to tender, with darkening of<br />

color to purple, peaking on the third day<br />

of hospitalization, affecting approximately<br />

70 percent of her body surface area.<br />

Biopsy revealed leukocytoclastic<br />

vasculitis, and the patient requested<br />

transfer for further management.<br />

On admission, corticosteroid was<br />

continued, since new onset granulomatosis<br />

with polyangiitis (Wegener’s) was<br />

suggested by the rash, nose deformity,<br />

proteinuria and recurrent sinusitis.<br />

Antibody to proteinase 3 was strongly<br />

positive. Review of her pathology<br />

slides deemed the underlying process<br />

to be classic for a cocaine-induced<br />

vasculitis. After an initial dose of pulse<br />

methylprednisolone, the patient was<br />

started on oral prednisone 1 mg/kg dose<br />

tapered over a 20-day period. There was<br />

very slow improvement in the cutaneous<br />

lesions, and she eventually underwent<br />

multiple sessions of skin grafting. At the<br />

time of discharge, there was marked<br />

improvement with mild scarring. Despite<br />

repeated questioning, the patient did not<br />

admit to cocaine use.<br />

Discussion<br />

Although we were unable to confirm that<br />

the presentation of this rash in our patient<br />

was related to cocaine use, the pathology<br />

report prompted us to understand the<br />

pathophysiology of cocaine-induced<br />

vasculitis. Unlike granulomatosis with<br />

polyangiitis, cocaine-induced vasculitis<br />

is less commonly a true vasculitis but<br />

rather a vasculopathy with thrombosis. Its<br />

occurrence is associated with cocaine that<br />

has been contaminated with levamisole,<br />

a veterinary antihelminthic agent, although<br />

recently it has been hypothesized that<br />

cocaine itself may enhance complement<br />

synthesis and trigger the formation of<br />

C5b-9, resulting in endothelial cell injury.<br />

While levamisole has been acknowledged as<br />

possessing immunomodulatory properties<br />

since the 1960s, the mechanism is not<br />

well understood. It reportedly potentiates<br />

the psychotropic effects of cocaine and<br />

therefore makes it highly desirable to<br />

manufacturers of illegal cocaine. <strong>Report</strong>s<br />

consistently state that it is added to<br />

approximately 70 percent of cocaine<br />

available in the United States.<br />

In cocaine-induced disease, the classic<br />

skin finding is that of retiform purpura, a<br />

net-like pattern of irregular nonblanching,<br />

violaceous plaques. Skin biopsy shows a<br />

very distinctive intravascular occlusion<br />

without vasculitis, termed vasculopathy,<br />

primarily targeting capillaries and<br />

venules. Gross et al. found, in 16 patients,<br />

that 13 percent had a pure small vessel<br />

vasculitis, 40 percent thrombotic<br />

vasculopathy, and 47 percent vasculitis<br />

and thrombosis. [1]<br />

While ANCA positivity has been<br />

documented with both cocaine use<br />

and with levamisole use individually,<br />

normalization of ANCA and APL antibodies<br />

after levamisole exposure tended to take<br />

place within 2–14 months in Rongioletti’s<br />

study. [2] Given this patient’s history,<br />

repeat serologies at 6–12 months will<br />

be useful.<br />

References<br />

1. Gross RL, et al. A novel cutaneous<br />

vasculitis syndrome induced by levamisolecontaminated<br />

cocaine. ClinRheumatol 2011;<br />

30(10):1385.<br />

2. Rongioletti F, et al. Purpura of the ears: a<br />

distinctive vasculopathy with circulating<br />

autoantibodies complicating long-term<br />

treatment with levamisole in children. Br J<br />

Dermatol 1999;140:948–951.<br />

Author Disclosures<br />

Dr. Danielle Ramsden-Stein does not have<br />

a financial interest or relationship with the<br />

manufacturers of products or services.<br />

Dr. Sonali Narain does not have a financial<br />

interest or relationship with the manufacturers<br />

of products or services.<br />

Dr. Michael D. Lockshin does not have a financial<br />

interest or relationship with the manufacturers<br />

of products or services.<br />

Figure 1: Extensive retiform purpuric lesions on<br />

patient’s bilateral lower extremities, Day 5.<br />

Figure 2: Saddle nose deformity. Resolving<br />

retiform purpura on cheeks.<br />

Figure 3: Skin biopsy shows a very distinctive<br />

intravascular occlusion without vasculitis primarily<br />

targeting capillaries and venules.


24<br />

Division of <strong>Rheumatology</strong><br />

<strong>HSS</strong> staff members, including Jillian Rose, LCSW, MPH, Director of Community Engagement, Diversity and<br />

Research (center), and Lillian Mendez, Program Associate, Charla de Lupus (Lupus Chat®) (second from right),<br />

with members of Charla de Lupus (Lupus Chat®) during the “Walk With Us to Cure Lupus” fundraiser, which<br />

benefits the Lupus Research Alliance.<br />

Empowering Patients Through<br />

Support and Education Programs<br />

At <strong>HSS</strong>, providing patient<br />

support and education is a key<br />

part of our mission to deliver<br />

the highest quality care. We<br />

offer a variety of services<br />

for patients with rheumatic<br />

diseases; in fact, we provide<br />

more support and education<br />

programs for lupus patients<br />

than any other hospital in the<br />

country. “Not only do we have<br />

the most programs, but we’ve<br />

been doing this the longest—for<br />

over 25 years,” says Stephen<br />

A. Paget, MD, Attending<br />

Physician and Physician-in-<br />

Chief Emeritus.<br />

<strong>HSS</strong> patients can learn more<br />

about their conditions in<br />

several different ways. Our<br />

physicians educate patients<br />

through Facebook Live events,<br />

video chats and in-person<br />

support group meetings.<br />

In addition, several of our<br />

Centers of Excellence offer<br />

webinars. Over the past year,<br />

the Inflammatory Arthritis<br />

Center of Excellence developed<br />

three educational webinars<br />

for patients in collaboration<br />

with the <strong>HSS</strong> Department of<br />

Social Work Programs, the <strong>HSS</strong><br />

Education Institute, the Arthritis<br />

Foundation and the Spondylitis<br />

Association of America. The<br />

Scleroderma, Vasculitis &<br />

Myositis Center of Excellence<br />

at <strong>HSS</strong> delivers a monthly email<br />

news blast to patients, and<br />

the <strong>HSS</strong> Division of Pediatric<br />

<strong>Rheumatology</strong> has developed<br />

patient education materials in<br />

multiple languages.<br />

Patient support and education<br />

programs are integral to the<br />

mission of the Department<br />

of Social Work Programs<br />

at <strong>HSS</strong>. “Our department<br />

partners with <strong>HSS</strong> physicians<br />

and healthcare teams to help<br />

support, empower and enhance<br />

the quality of life for people<br />

with rheumatologic conditions,”<br />

says Roberta Horton, LCSW,<br />

ACSW, Assistant Vice President,<br />

Department of Social Work<br />

Programs. <strong>HSS</strong> offers programs<br />

for patients with specific<br />

rheumatologic conditions,<br />

including the following:


Hospital for Special Surgery 25<br />

Lupus<br />

SLE Workshop<br />

This support and education program for lupus<br />

patients and their families and friends has been<br />

ongoing since 1985. Lupus care experts give<br />

lectures, which are followed by informal discussions.<br />

LANtern® (Lupus Asian Network)<br />

LANtern is the only national, hospital-based<br />

bilingual support and education program<br />

dedicated to serving Asian Americans with lupus<br />

and their families. Asians are more likely to develop<br />

lupus than Caucasian people, and their illness<br />

may be more severe. LANtern offers a bilingual,<br />

toll-free telephone support line of peer educators.<br />

These trained volunteers—either Asian Americans<br />

with lupus or Asian Americans who have family<br />

members with lupus—can help callers better<br />

understand their condition, address any concerns,<br />

and offer advice on how to work effectively with<br />

their physicians.<br />

Charla de Lupus (Lupus Chat®)<br />

This unique national program provides peer<br />

support and education to English- and Spanishspeaking<br />

people with lupus. It includes the Charla<br />

Line, a toll-free support and education helpline<br />

that screens and matches callers with peers.<br />

In addition, the program offers peer support at<br />

several rheumatology clinics in New York City.<br />

“Since 1988, we’ve trained lupus patients to<br />

provide specific support to families while they<br />

are sitting in the waiting room,” says Jillian Rose,<br />

LCSW, MPH, Director of Community Engagement,<br />

Diversity and Research. Program staff members<br />

conduct more than 30 community and professional<br />

presentations annually to raise awareness of lupus<br />

and provide access to educational resources.<br />

Additional programs offer support to teens and<br />

their parents.<br />

Rheumatoid Arthritis<br />

Living with Rheumatoid Arthritis provides<br />

monthly education and support for patients with<br />

moderate to severe rheumatoid arthritis (RA) and<br />

their families. RA care experts give lectures, which<br />

are followed by a discussion facilitated by a clinical<br />

social worker and rheumatology nurse.<br />

The Early RA Support and Education Program was<br />

developed for people who were recently diagnosed<br />

with RA. Participants meet for a free, four-session<br />

workshop series, which provides tips on how to<br />

communicate with their physician about RA and<br />

manage activities of daily living.<br />

Myositis<br />

<strong>HSS</strong> sponsors a Myositis Support Group, and meetings are generally held once a month at the<br />

Hospital. During the first hour, speakers such as myositis specialists, social workers and patients give<br />

presentations on topics as varied as managing your medical team, fatigue, and current research and<br />

treatments. During the second hour, members are able to share their thoughts and feelings about<br />

the illness.<br />

Scleroderma<br />

<strong>HSS</strong> hosts a monthly scleroderma support group sponsored by the Tri-State Chapter of the Scleroderma<br />

Foundation. In addition, the Scleroderma, Vasculitis & Myositis Center of Excellence hosts two<br />

Scleroderma Foundation patient forums every year.<br />

For more information on these programs, visit hss.edu/support-programs<br />

Using technology to assist lupus patients<br />

In <strong>2017</strong>, <strong>HSS</strong> launched LupusMinder, a mobile app designed to help lupus patients track medications,<br />

daily symptoms and appointments. Unlike other apps, this one was created with significant input from<br />

lupus patients themselves. The app enables users to take photos of their symptoms to share with their<br />

physician and chart daily symptoms, such as pain.


26<br />

Division of <strong>Rheumatology</strong><br />

<strong>Rheumatology</strong><br />

Leaders<br />

of the Future<br />

Bella Mehta, MBBS, MD<br />

Iris Navarro-Millán, MD, MSPH<br />

Medha Barbhaiya, MD, MPH<br />

<strong>Rheumatology</strong> has always attracted physicians who<br />

enjoy problem solving and managing some of the most<br />

complex cases in medicine. This is especially evident<br />

among the newest rheumatologists at <strong>HSS</strong>, who are<br />

committed to performing cutting-edge research and<br />

revolutionizing patient care. Here are their hopes for<br />

the future of the field.<br />

David R. Fernandez, MD, PhD


Hospital for Special Surgery 27<br />

Medha Barbhaiya, MD, MPH<br />

Medha Barbhaiya, MD, MPH, Assistant Attending Physician, is developing<br />

new classification criteria for APS and researching environmental risk<br />

factors for lupus.<br />

Medha Barbhaiya, MD, MPH,<br />

Assistant Attending Physician,<br />

is poised to become a leading<br />

researcher on lupus and<br />

Antiphospholipid Syndrome<br />

(APS). In 2015, Dr. Barbhaiya<br />

received the <strong>Rheumatology</strong><br />

Research Scientist Development<br />

Award to study environmental<br />

risk factors for lupus. “One of<br />

the biggest mysteries is why<br />

lupus seems to affect young,<br />

previously healthy patients,”<br />

she says. “Some patients may<br />

have an underlying genetic<br />

predisposition, but we hope<br />

to identify additional factors<br />

that contribute to their risk of<br />

developing lupus.” Dr. Barbhaiya<br />

studies lupus risk factors in<br />

over 230,000 participants in<br />

the Nurses’ Health Study and<br />

59,000 participants in the<br />

Black Women’s Health Study.<br />

She is examining the role of<br />

smoking, alcohol intake, diet<br />

(including the Mediterranean,<br />

Alternative Healthy Eating<br />

Index, and Inflammatory diets),<br />

UV light exposure, and even<br />

psychological factors, such<br />

as child abuse. In a study<br />

published in <strong>2018</strong> in Annals<br />

of the Rheumatic Diseases,<br />

Dr. Barbhaiya found that while<br />

cigarette smoking was not<br />

associated with a greater risk<br />

of developing lupus overall,<br />

it was for a subset of lupus<br />

patients who had a specific<br />

type of antibody known as antidouble<br />

stranded DNA (dsDNA).<br />

In another study published in<br />

August <strong>2018</strong> in Arthritis Care<br />

& Research, she demonstrated<br />

that while smoking slightly<br />

elevated the risk of developing<br />

lupus, moderate alcohol<br />

intake seemed protective. “In<br />

moderate amounts, alcohol<br />

may have an anti-inflammatory<br />

effect,” she says.<br />

In addition to her ongoing<br />

lupus-related research studies,<br />

Dr. Barbhaiya has been involved<br />

in APS research since medical<br />

school. She received a “Young<br />

Scholar” award from the APS<br />

Alliance for Clinical Trials<br />

and International Networking<br />

(APS ACTION) in 2014. In<br />

<strong>2018</strong>, she was the recipient<br />

of an Investigator Award from<br />

the <strong>Rheumatology</strong> Research<br />

Foundation to support her<br />

APS research. Currently, she<br />

is investigating risk factors<br />

for the development of<br />

thrombosis in APS in a spinoff<br />

study of an ACR/EULARsupported<br />

multidisciplinary<br />

effort to develop new APS<br />

classification criteria, for which<br />

she is one of the core planning<br />

group members. Studies of<br />

APS have been limited by<br />

small sample sizes, varying<br />

definitions of the disease,<br />

and laboratory tests that are<br />

not standardized. “There’s an<br />

urgent need for rigorous, highperforming<br />

APS classification<br />

criteria,” says Dr. Barbhaiya.<br />

As part of the classification<br />

criteria development efforts,<br />

Dr. Barbhaiya has been working<br />

with a large, international group<br />

of APS researchers, including<br />

other specialists, such as<br />

hematologists, pathologists and<br />

dermatologists. She plans to<br />

use international, multicenter<br />

patient data to investigate<br />

associations between potentially<br />

modifiable environmental<br />

factors and the risk of<br />

developing APS. Ultimately,<br />

she hopes that identifying risk<br />

factors will lead to improved<br />

prevention strategies.<br />

“For lupus and APS, there are so<br />

many unanswered questions,”<br />

says Dr. Barbhaiya. “Hopefully,<br />

I can help answer some of<br />

these questions and see the<br />

impact of my research on my<br />

patients’ care.” There is no<br />

better institution to support<br />

this work than <strong>HSS</strong>, since<br />

the Hospital takes a multidisciplinary<br />

approach to lupus<br />

and APS, says Dr. Barbhaiya.<br />

“There’s a lot of camaraderie<br />

and synergy between clinicians<br />

and researchers,” she says.<br />

“There’s also a tremendous<br />

amount of support for junior<br />

faculty. We are given time to<br />

perform research while serving<br />

as clinicians.”


28<br />

Division of <strong>Rheumatology</strong><br />

David R. Fernandez, MD, PhD<br />

David R. Fernandez, MD, PhD, Assistant Attending Physician, aims to study the molecular mechanisms that<br />

drive myositis to develop better treatments.<br />

David R. Fernandez, MD, PhD,<br />

Assistant Attending Physician,<br />

has dedicated his career to<br />

researching and caring for<br />

patients with myositis. While he<br />

has successfully treated many,<br />

he’s haunted by thoughts of<br />

those who haven’t responded to<br />

treatment. “This provides strong<br />

motivation to try to help patients<br />

further,” says Dr. Fernandez,<br />

who is researching the causes<br />

and treatment of myositis.<br />

“Uncommon disorders like these<br />

could use more support, and<br />

<strong>HSS</strong> is in a great position to<br />

provide that.”<br />

Dr. Fernandez is creating a<br />

registry of myositis patients to<br />

study the natural history of the<br />

disease. “At <strong>HSS</strong>, we see a high<br />

volume of patients, which helps<br />

to inform research and move the<br />

field forward,” he says. Currently,<br />

Dr. Fernandez is collecting blood<br />

samples to determine whether<br />

T cells and B cells in myositis<br />

patients have some of the<br />

same abnormalities commonly<br />

found in lupus patients. “There<br />

are a lot of clinical similarities<br />

between the two diseases, so<br />

we’re hoping we may be able<br />

to confirm that on a molecular<br />

level,” he explains. Dr. Fernandez<br />

is also participating in a clinical<br />

trial focusing on the effects<br />

of the lupus drug belimumab<br />

to treat dermatomyositis and<br />

polymyositis. He is studying the<br />

protein mTOR, which is altered<br />

in people with lupus. “One recent<br />

clinical trial found that a drug<br />

that targeted mTOR showed<br />

some benefit in lupus patients,<br />

so we’d like to see if this protein<br />

is also affected in myositis,<br />

which may indicate similar<br />

therapies might help some of our<br />

myositis patients,” he explains.<br />

Dr. Fernandez is the physician<br />

advisor to the <strong>HSS</strong> Myositis<br />

Support and Education Program.<br />

He attends support group<br />

meetings and speaks to patients<br />

about the latest advances in<br />

treatment and research. “I spend<br />

80 percent of my time doing<br />

research, but the 20 percent<br />

of time spent with patients is<br />

invaluable,” he says. “It reminds<br />

me of the acuity of these<br />

illnesses and how important it<br />

is to move the field forward. It<br />

also benefits me as a researcher<br />

because it helps me understand<br />

how much these diseases can<br />

vary from person to person.”<br />

Ultimately, Dr. Fernandez<br />

hopes to run his own myositis<br />

laboratory at <strong>HSS</strong>, which would<br />

allow him to study the molecular<br />

mechanisms that drive the<br />

disease. “I have many colleagues<br />

with expertise in different<br />

diseases,” he says. “If I’m having<br />

trouble with a case history or<br />

designing a study protocol,<br />

they’re just down the hall. I can’t<br />

imagine a better environment to<br />

do research and treat patients.”


Hospital for Special Surgery 29<br />

Bella Mehta, MBBS, MD<br />

Bella Mehta, MBBS, MD,<br />

Assistant Attending Physician,<br />

became interested in the<br />

field of rheumatology when<br />

she came to <strong>HSS</strong> in 2010 as<br />

a visiting medical student<br />

from India. One of her first<br />

cases involved diagnosing a<br />

patient who had suffered from<br />

persistent, unexplained fevers<br />

for many years. It took multiple<br />

specialists to come up with a<br />

diagnosis: adult-onset Still’s<br />

disease, a rare autoimmune<br />

disorder. “<strong>Rheumatology</strong> cases<br />

are never straightforward<br />

and simple,” says Dr. Mehta.<br />

“There’s something very<br />

satisfying about making the<br />

right diagnosis.”<br />

Today, Dr. Mehta is putting her<br />

sleuthing skills to good use,<br />

analyzing <strong>HSS</strong> registries and large<br />

data sets to address important<br />

questions about healthcare<br />

disparities and outcomes for<br />

patients with arthritis. Using<br />

geospatial epidemiology, she<br />

and several colleagues, including<br />

Susan M. Goodman, MD, Director<br />

of the Integrative <strong>Rheumatology</strong><br />

and Orthopedic Center of<br />

Excellence and Attending<br />

Physician, investigated how<br />

neighborhood characteristics—<br />

specifically the proportion<br />

of impoverished people and<br />

immigrants—influence outcomes<br />

after joint replacement surgery at<br />

<strong>HSS</strong>. “Geospatial epidemiology is<br />

usually thought of as a marketing<br />

tool (businesses use it in different<br />

neighborhoods to figure out what<br />

types of food residents eat),<br />

but it can help us understand<br />

how where a person lives affects<br />

health outcomes.”<br />

While pain and function scores<br />

after total hip replacement<br />

were similar for blacks<br />

and whites in middle-class<br />

or affluent communities,<br />

Bella Mehta, MBBS, MD, Assistant Attending Physician, is investigating<br />

disparities in healthcare among arthritis patients.<br />

Dr. Mehta found that blacks<br />

in poorer neighborhoods<br />

fared significantly worse than<br />

their white counterparts. In<br />

other research, Dr. Mehta<br />

discovered that education can<br />

help mitigate the effects of<br />

these outcomes. “In poorer<br />

communities, those who have<br />

some college education have<br />

significantly better pain and<br />

function scores than those who<br />

don’t,” she says. These findings<br />

suggest that patients without<br />

a college education who live in<br />

economically disadvantaged<br />

neighborhoods should be<br />

targeted for preoperative and<br />

postoperative interventions,<br />

including being encouraged<br />

to have procedures like joint<br />

replacement earlier to achieve<br />

better outcomes.<br />

In <strong>2017</strong>, Dr. Mehta’s work on<br />

epidemiology and rheumatology<br />

outcomes was selected as one<br />

of the top three research studies<br />

at the American College of<br />

<strong>Rheumatology</strong> annual meeting.<br />

Dr. Mehta’s goal is to decrease<br />

disparities, reduce disability and<br />

improve outcomes in arthritis<br />

patients. “It’s a different level<br />

of disparity than what I was<br />

used to in India, where many<br />

impoverished people simply go<br />

without health care because<br />

resources such as Medicare aren’t<br />

available,” she says. “In the U.S.,<br />

even if someone has Medicare<br />

or Medicaid, they still might not<br />

have surgery or see their primary<br />

care physician. The question<br />

becomes, what other factors are<br />

holding people back, and why?”<br />

Dr. Mehta is confident she will be<br />

able to answer these questions at<br />

<strong>HSS</strong>. “The intellectual stimulation<br />

here is so energizing,” she says.<br />

“We have some of the greatest<br />

minds in medicine here.”


30<br />

Division of <strong>Rheumatology</strong><br />

Iris Navarro-Millán, MD, MSPH<br />

Iris Navarro-Millán, MD, MSPH, Assistant Attending Physician, is<br />

studying the prevalence and incidence of cardiovascular risk factors<br />

among patients with psoriatic and rheumatoid arthritis.<br />

For more than half a century,<br />

research has shown that<br />

rheumatoid arthritis (RA)<br />

raises the risk of developing<br />

cardiovascular disease (CVD).<br />

Despite this, many physicians<br />

and patients are unaware of<br />

the connection: One recent<br />

study found that about half of<br />

all patients with RA and their<br />

primary care providers were<br />

unaware of the link and were<br />

not taking steps to implement<br />

preventive care. Iris Navarro-<br />

Millán, MD, MSPH, Assistant<br />

Attending Physician, is<br />

attempting to close that gap.<br />

Over the last few years, she has<br />

been studying the prevalence<br />

and incidence of cardiovascular<br />

risk factors—high blood<br />

pressure, elevated cholesterol<br />

levels, obesity and Type 2<br />

diabetes—among patients<br />

with psoriatic and rheumatoid<br />

arthritis. She published a study<br />

in Annals of the Rheumatic<br />

Diseases highlighting the<br />

association of high levels of<br />

inflammatory markers, such as<br />

C-reactive protein (CRP) and<br />

erythrocyte sedimentation rate<br />

(ESR), with an increased risk of<br />

heart attack and stroke among<br />

veterans with RA. “Our research<br />

shows that this inflammation<br />

is driving the increase in<br />

cardiovascular disease among<br />

people with rheumatoid<br />

arthritis,” she explains.<br />

Managing cardiovascular<br />

risk factors in RA patients<br />

is key, yet a study published<br />

by Dr. Navarro-Millán in the<br />

Journal of International Medical<br />

Research in <strong>2018</strong> found that<br />

patients with inflammatory<br />

arthritis (rheumatoid arthritis,<br />

psoriatic arthritis and ankylosing<br />

spondylitis) were no more<br />

likely to be treated for high<br />

cholesterol than people without<br />

any cardiovascular risk factors.<br />

In her most recent study, she<br />

found that only 37 percent of<br />

patients with RA are screened<br />

for hyperlipidemia. This article<br />

was published in November <strong>2018</strong><br />

in Arthritis Care and Research,<br />

and it was highlighted as one<br />

of the must-read articles for<br />

the first week of December by<br />

<strong>Rheumatology</strong> News Clinical<br />

Edge. As a result, Dr. Navarro-<br />

Millán is currently developing<br />

an intervention to address the<br />

gaps in patient care. “We’re<br />

focusing mainly on improving<br />

screening and management<br />

of hyperlipidemia among<br />

patients with RA,” she says.<br />

“We’re developing a series of<br />

brief videos for patients that<br />

explain the short- and long-term<br />

effects of RA and medications.<br />

The videos will also cover the<br />

importance of screening and<br />

management of cholesterol<br />

levels as a way of decreasing<br />

their risk of complications.” The<br />

videos will be available online<br />

for patients to watch on their<br />

own, but they can also receive<br />

the assistance of a peer coach<br />

to guide them. Peer coaches<br />

are individuals who have RA<br />

and have received training to<br />

provide guidance regarding the<br />

video content and support. “We<br />

do not anticipate that everyone<br />

with RA will need a peer coach,<br />

but this option will be available<br />

for those individuals who have<br />

limited experience navigating<br />

the healthcare system and may<br />

need guidance and support to<br />

understand the importance of<br />

CVD risk as part of having RA,”<br />

says Dr. Navarro-Millán. “We<br />

aim to make these resources<br />

accessible to everyone in<br />

this country.”<br />

Dr. Navarro-Millán is confident<br />

that the community of physicianresearchers<br />

at <strong>HSS</strong> will help her<br />

achieve her goal of improving<br />

the care of RA patients. “Having<br />

such a diverse cadre of clinicians<br />

to collaborate with and to help<br />

me generate questions I can<br />

develop into meaningful clinical<br />

research studies is, for me, the<br />

biggest asset of working at <strong>HSS</strong>,”<br />

she says.


Hospital for Special Surgery 31<br />

Notable References<br />

Ah Kioon MD, Tripodo C, Fernandez D, Kirou KA, Spiera RF,<br />

Crow MK, Gordon JK, Barrat FJ. Plasmacytoid dendritic<br />

cells promote systemic sclerosis with a key role for TLR8.<br />

Sci Med. <strong>2018</strong> Jan 10;10(423).<br />

Bass AR, Fields KG, Goto R, Turissini G, Dey S, Russell<br />

LA. Clinical Decision Rules for Pulmonary Embolism<br />

in Hospitalized Patients: A Systematic Literature<br />

Review and Meta-analysis. Thromb Haemost. <strong>2017</strong><br />

Nov;117(11):2176–2185.<br />

Brunner HI, Rider LG, Kingsbury DJ, Co D, Schneider R,<br />

Goldmuntz E, Onel KB, Giannini EH, Lovell DJ; PRCSG<br />

Advisory Council. Pediatric <strong>Rheumatology</strong> Collaborative<br />

Study Group—over four decades of pivotal clinical drug<br />

research in pediatric rheumatology. Pediatr Rheumatol<br />

Online J. <strong>2018</strong> Jul 11;16(1):45.<br />

Chan K, Bass AR. Checkpoint inhibitor-induced<br />

polymyalgia rheumatica controlled by cobimetinib, a<br />

MEK 1/2 inhibitor. Annals of the Rheumatic Diseases.<br />

May <strong>2018</strong>, annrheumdis-<strong>2018</strong>-213672; doi: 10.1136/<br />

annrheumdis-<strong>2018</strong>-213672.<br />

Donlin LT, Rao DA, Wei K, Slowikowski K, McGeachy MJ,<br />

Turner JD, Meednu N, Mizoguchi F, Gutierrez-Arcelus M,<br />

Lieb DJ, Keegan J, Muskat K, Hillman J, Rozo C, Ricker<br />

E, Eisenhaure TM, Li S, Browne EP, Chicoine A, Sutherby<br />

D, Noma A; Accelerating Medicines Partnership RA/<br />

SLE Network, Nusbaum C, Kelly S, Pernis AB, Ivashkiv<br />

LB, Goodman SM, Robinson WH, Utz PJ, Lederer JA,<br />

Gravallese EM, Boyce BF, Hacohen N, Pitzalis C, Gregersen<br />

PK, Firestein GS, Raychaudhuri S, Moreland LW, Holers<br />

VM, Bykerk VP, Filer A, Boyle DL, Brenner MB, Anolik JH.<br />

Methods for high-dimensional analysis of cells dissociated<br />

from cyropreserved synovial tissue. Arthritis Res Ther.<br />

<strong>2018</strong> Jul 11;20(1):139.<br />

Fields TR, Batterman A. How Can We Improve Disease<br />

Education in People with Gout? Curr Rheumatol Rep. <strong>2018</strong><br />

Mar 8;20(3):12.<br />

Fu MC, Boddapati V, Gausden EB, Samuel AM, Russell LA,<br />

Lane JM. Surgery for a fracture of the hip within 24 hours<br />

of admission is independently associated with reduced<br />

short-term post-operative complications. Bone Joint J.<br />

<strong>2017</strong> Sep;99-B(9):1216–1222.<br />

Gage BF, Bass AR, Lin H, Woller SC, Stevens SM, Al-<br />

Hammadi N, Li J, Rodríguez T Jr, Miller JP, McMillin<br />

GA, Pendleton RC, Jaffer AK, King CR, Whipple BD,<br />

Porche-Sorbet R, Napoli L, Merritt K, Thompson AM,<br />

Hyun G, Anderson JL, Hollomon W, Barrack RL, Nunley<br />

RM, Moskowitz G, Dávila-Román V, Eby CS. Effect of<br />

Genotype-Guided Warfarin Dosing on Clinical Events and<br />

Anticoagulation Control Among Patients Undergoing Hip<br />

or Knee Arthroplasty: The GIFT Randomized Clinical Trial.<br />

JAMA. <strong>2017</strong> Sep 26;318(12):1115–1124.<br />

Garcia D, Erkan D. Diagnosis and Management of the<br />

Antiphospholipid Syndrome. N Engl J Med. <strong>2018</strong> May<br />

24;378(21):2010–2021.<br />

Goodman SM, Bykerk VP, DiCarlo E, Cummings RW, Donlin<br />

LT, Orange DE, Hoang A, Mirza S, McNamara M, Andersen<br />

K, Bartlett SJ, Szymonifka J, Figgie MP. Flares in Patients<br />

with Rheumatoid Arthritis after Total Hip and Total Knee<br />

Arthroplasty: Rates, Characteristics, and Risk Factors.<br />

J Rheumatol. <strong>2018</strong> May;45(5):604–611.<br />

Goodman SM, Mandl LA, Mehta B, Navarro-Millán I, Russell<br />

LA, Parks ML, Dey SA, Crego D, Figgie MP, Nguyen JT,<br />

Szymonifka J, Zhang M, Bass AR. Does Education Level<br />

Mitigate the Effect of Poverty on Total Knee Arthroplasty<br />

Outcomes? Arthritis Care Res (Hoboken). <strong>2018</strong><br />

Jun;70(6):884–891.<br />

Goodman SM, Springer B, Guyatt G, Abdel MP, Dasa V,<br />

George M, Gewurz-Singer O, Giles JT, Johnson B, Lee<br />

S, Mandl LA, Mont MA, Sculco P, Sporer S, Stryker L,<br />

Turgunbaev M, Brause B, Chen AF, Gililland J, Goodman<br />

M, Hurley-Rosenblatt A, Kirou K, Losina E, MacKenzie R,<br />

Michaud K, Mikuls T, Russell L, Sah A, Miller AS, Singh<br />

JA, Yates A. <strong>2017</strong> American College of <strong>Rheumatology</strong>/<br />

American Association of Hip and Knee Surgeons Guideline<br />

for the Perioperative Management of Antirheumatic<br />

Medication in Patients With Rheumatic Diseases<br />

Undergoing Elective Total Hip or Total Knee Arthroplasty.<br />

J Arthroplasty. <strong>2017</strong> Sep;32(9):2628–2638.<br />

Gordon JK, Martyanov V, Franks JM, Bernstein EJ,<br />

Szymonifka J, Magro C, Wildman HF, Wood TA, Whitfield<br />

ML, Spiera RF. Belimumab for the Treatment of Early<br />

Diffuse Systemic Sclerosis: Results of a Randomized,<br />

Double-Blind, Placebo-Controlled, Pilot Trial. Arthritis<br />

Rheumatol. <strong>2018</strong> Feb;70(2):308–316.<br />

Horton DB, Onel KB, Beukelman T, Ringold S. Attitudes<br />

and Approaches for Withdrawing Drugs for Children<br />

with Clinically Inactive Nonsystemic JIA: A Survey of the<br />

Childhood Arthritis and <strong>Rheumatology</strong> Research Alliance.<br />

J Rheumatol. <strong>2017</strong> Mar;44(3):352–360.<br />

Kim MY, Guerra MM, Kaplowitz E, Laskin CA, Petri M,<br />

Branch DW, Lockshin MD, Sammaritano LR, Merrill JT,<br />

Porter TF, Sawitzke A, Lynch AM, Buyon JP, Salmon JE.<br />

Complement activation predicts adverse pregnancy<br />

outcome in patients with systemic lupus erythematosus<br />

and/or antiphospholipid antibodies. Ann Rheum Dis. <strong>2018</strong><br />

Apr;77(4):549-555.<br />

Lafyatis R, Mantero JC, Gordon J, Kishore N, Carns M,<br />

Dittrich H, Spiera R, Simms RW, Varga J. Inhibition of<br />

β-Catenin Signaling in the Skin Rescues Cutaneous<br />

Adipogenesis in Systemic Sclerosis: A Randomized,<br />

Double-Blind, Placebo-Controlled Trial of C-82. J Invest<br />

Dermatol. <strong>2017</strong> Dec;137(12):2473–2483.<br />

Liu Y, Carrino JA, Dash AS, Chukir T, Do H, Bockman RS,<br />

Hughes AP, Press JM, Stein EM. Lower spine volumetric<br />

bone density in patients with a history of epidural steroid<br />

injections. J Clin Endocrinol Metab. <strong>2018</strong> Jul 2. doi:<br />

10.1210/jc.<strong>2018</strong>-00558.


32<br />

Division of <strong>Rheumatology</strong><br />

Liu Y, Dimango E, Bucovsky M, Agarwal S, Nishiyama K,<br />

Guo XE, Shane E, Stein EM. Abnormal microarchitecture<br />

and stiffness in postmenopausal women using chronic<br />

inhaled glucocorticoids. Osteoporos Int. <strong>2018</strong> Jun 11. doi:<br />

10.1007/s00198-018-4591–9.<br />

Manni M, Gupta S, Ricker E, Chinenov Y, Park SH, Shi<br />

M, Pannellini T, Jessberger R, Ivashkiv LB, Pernis AB.<br />

Regulation of age-associated B cells by IRF5 in systemic<br />

autoimmunity. Nat Immunol. <strong>2018</strong> Apr;19(4):407–419.<br />

Marangoni RG, Lu TT. The roles of dermal white adipose<br />

tissue loss in scleroderma skin fibrosis. Curr Opin<br />

Rheumatol. <strong>2017</strong> Nov;29(6):585–590.<br />

Mavragani CP, Nezos A, Sagalovskiy I, Seshan S, Kirou<br />

KA, Crow MK. Defective regulation of L1 endogenous<br />

retroelements in primary Sjogren’s syndrome and systemic<br />

lupus erythematosus: Role of methylating enzymes.<br />

J Autoimmun. <strong>2018</strong> Mar;88:75–82.<br />

Mehta BY, Bass AR, Goto R, Russell LA, Parks ML, Figgie<br />

MP, Goodman SM. Disparities in Outcomes for Blacks<br />

versus Whites Undergoing Total Hip Arthroplasty:<br />

A Systematic Literature Review. J Rheumatol. <strong>2018</strong><br />

May;45(5):717–722.<br />

Navarro-Millán I, Zinski A, Shurbaji S, Johnson B,<br />

Fraenkel L, Willig J, Danila MI, Yun H, Curtis JR, Safford<br />

MM. Perspectives of Rheumatoid Arthritis Patients on<br />

Electronic Communication and Patient <strong>Report</strong>ed Outcome<br />

Data Collection: A Qualitative Study. Arthritis Care Res<br />

(Hoboken). <strong>2018</strong> Apr 18. doi: 10.1002/acr.23580.<br />

Orange DE, Agius P, DiCarlo EF, Robine N, Geiger H,<br />

Szymonifka J, McNamara M, Cummings R, Andersen KM,<br />

Mirza S, Figgie M, Ivashkiv LB, Pernis AB, Jiang CS, Frank<br />

MO, Darnell RB, Lingampali N, Robinson WH, Gravallese<br />

E; Accelerating Medicines Partnership in Rheumatoid<br />

Arthritis and Lupus Network, Bykerk VP, Goodman SM,<br />

Donlin LT. Identification of Three Rheumatoid Arthritis<br />

Disease Subtypes by Machine Learning Integration of<br />

Synovial Histologic Features and RNA Sequencing Data.<br />

Arthritis Rheumatol. <strong>2018</strong> May;70(5):690–701.<br />

Park SH, Kang K, Giannopoulou E, Qiao Y, Kang K, Kim<br />

G, Park-Min KH, Ivashkiv LB. Type I interferons and the<br />

cytokine TNF cooperatively reprogram the macrophage<br />

epigenome to promote inflammatory activation. Nat<br />

Immunol. <strong>2017</strong> Oct;18(10):1104–1116.<br />

Park-Min KH. Mechanisms involved in normal and<br />

pathological osteoclastogenesis. Cell Mol Life Sci. <strong>2018</strong><br />

Jul;75(14):2519–2528.<br />

Qing X, Chinenov Y, Redecha P, Madaio M, Roelofs JJ,<br />

Farber G, Issuree PD, Donlin L, Mcllwain DR, Mak TW,<br />

Blobel CP, Salmon JE. iRhom2 promotes lupus nephritis<br />

through TNF-α and EGFR signaling. J Clin Invest. <strong>2018</strong><br />

Apr 2;128(4):1397–1412.<br />

Russell LA, Sigmund AE, Szymonifka J, Jawetz ST, Grond<br />

SE, Dey SA, Bass AR. Does my patient have a pulmonary<br />

embolism? The Wells vs. PISA 2 rule in orthopedic patients.<br />

J Thromb Thrombolysis. <strong>2018</strong> Apr;45(3):417–422.<br />

Schulman E, Bartlett SJ, Schieir O, Andersen KM,<br />

Boire G, Pope JE, Hitchon C, Jamal S, Thorne JC, Tin D,<br />

Keystone EC, Haraoui B, Goodman SM, Bykerk VP; CATCH<br />

Investigators. Overweight, Obesity, and the Likelihood<br />

of Achieving Sustained Remission in Early Rheumatoid<br />

Arthritis: Results From a Multicenter Prospective Cohort<br />

Study. Arthritis Care Res (Hoboken). <strong>2017</strong> Nov 30. doi:<br />

10.1002/acr.23457.<br />

Shipman WD, Chyou S, Ramanathan A, Izmirly PM, Sharma<br />

S, Pannellini T, Dasoveanu DC, Qing X, Magro CM, Granstein<br />

RD, Lowes MA, Pamer EG, Kaplan DH, Salmon JE, Mehrara<br />

BJ, Young JW, Clancy RM, Blobel CP, Lu TT. A prospective<br />

Langerhans cell-keratinocyte axis that is dysfunctional in<br />

photosensitivity. Sci Transl Med. <strong>2018</strong> Aug 15;10(454).<br />

Smith MH, Bass AR. Arthritis after cancer immunotherapy:<br />

symptom duration and treatment response. Arthritis Care<br />

Res (Hoboken). <strong>2017</strong> Nov 10.<br />

Stephenson W, Donlin LT, Butler A, Rozo C, Bracken B,<br />

Rashidfarrokhi A, Goodman SM, Ivashkiv LB, Bykerk VP,<br />

Orange DE, Darnell RB, Swerdlow HP, Satija R. Single-cell<br />

RNA-seq of rheumatoid arthritis synovial tissue using lowcost<br />

microfluidic instrumentation. Nat Commun. <strong>2018</strong> Feb<br />

23;9(1):791.<br />

Stone JH, Tuckwell K, Dimonaco S, Klearman M, Aringer<br />

M, Blockmans D, Brouwer E, Cid MC, Bhaskar D, Rech J,<br />

Salvarani C, Schett G, Schulze-Koops H, Spiera R, Unizony<br />

SH, Collinson N. Trial of Tocilizumab in Giant-Cell Arteritis.<br />

N Engl J Med. <strong>2017</strong>;377(4):317–328.<br />

Wei W, Knapp K, Wang L, Chen CI, Craig GL, Ferguson<br />

K, Schwartzman S. Treatment Persistence and Clinical<br />

Outcomes of Tumor Necrosis Factor Inhibitor Cycling<br />

or Switching to a New Mechanism of Action Therapy:<br />

Real-world Observational Study of Rheumatoid<br />

Arthritis Patients in the United States with Prior Tumor<br />

Necrosis Factor Inhibitor Therapy. Adv Ther. <strong>2017</strong><br />

Aug;34(8):1936–1952.<br />

Yi W, Gupta S, Ricker E, Manni M, Jessberger R, Chinenov<br />

Y, Molina H, Pernis AB. The mTORC1-4E-BP-eIF4E<br />

axis controls de novo Bcl6 protein synthesis in T cells<br />

and systemic autoimmunity. Nat Commun. <strong>2017</strong> Aug<br />

15;8(1):254.<br />

Zhao Y, Wu EY, Oliver MS, Cooper AM, Basiaga ML, Vora SS,<br />

Lee TC, Fox E, Amarilyo G, Stern SM, Dvergsten JA, Haines<br />

KA, Rouster-Stevens KA, Onel KB, Cherian J, Hausmann<br />

JS, Miettunen P, Cellucci T, Nuruzzaman F, Taneja A, Barron<br />

KS, Hollander MC, Lapidus SK, Li SC, Ozen S, Girschick H,<br />

Laxer RM, Dedeoglu F, Hedrich CM, Ferguson PJ; Chronic<br />

Nonbacterial Osteomyelitis/Chronic Recurrent Multifocal<br />

Osteomyelitis Study Group and the Childhood Arthritis and<br />

<strong>Rheumatology</strong> Research Alliance Scleroderma, Vasculitis,<br />

Autoinflammatory and Rare Diseases Subcommittee.<br />

Consensus Treatment Plans for Chronic Nonbacterial<br />

Osteomyelitis Refractory to Nonsteroidal Antiinflammatory<br />

Drugs and/or With Active Spinal Lesions. Arthritis Care Res<br />

(Hoboken). <strong>2017</strong> Nov 7. doi: 10.1002/acr.23462


About Hospital for Special Surgery<br />

Founded in 1863, Hospital for Special Surgery (<strong>HSS</strong>) is a world leader in orthopaedics, rheumatology and rehabilitation.<br />

<strong>HSS</strong> is ranked #1 in the U.S. for Orthopaedics and #3 for <strong>Rheumatology</strong> by U.S. News & World <strong>Report</strong> “Best Hospitals”<br />

(<strong>2018</strong>–2019 rankings). It is also ranked Best in New York City for Pediatric Orthopaedics and #21 nationally by U.S.<br />

News & World <strong>Report</strong> “Best Hospitals” (<strong>2018</strong>–2019 rankings). It is the first hospital in New York State to receive the<br />

Magnet® designation for Excellence in Nursing Service from the American Nurses Credentialing Center four consecutive<br />

times. Located in New York City, <strong>HSS</strong> also serves patients in the regional area with outpatient centers in Westchester<br />

County, New York; Connecticut; New Jersey; Long Island and Queens. In 2019, there will be a new <strong>HSS</strong> location in West<br />

Palm Beach, FL. Patients choose to come to <strong>HSS</strong> from across the U.S. and from around the world. <strong>HSS</strong> has one of the<br />

lowest infection rates in the country. The Hospital’s Research Institute is internationally recognized as a leader in the<br />

investigation of musculoskeletal and autoimmune diseases. To learn more, please visit hss.edu.<br />

Attributions<br />

The <strong>HSS</strong> Division of <strong>Rheumatology</strong> <strong>Annual</strong> <strong>Report</strong> is published by the Communications Department, Hospital for Special<br />

Surgery, 535 East 70th Street, New York, NY 10021. 866-976-1196<br />

Hospital for Special Surgery is an affiliate of Weill Cornell Medical College.<br />

©<strong>2018</strong> Hospital for Special Surgery. All rights reserved.<br />

Officers<br />

Co-Chairs<br />

Thomas Lister<br />

Robert K. Steel<br />

Vice Chair<br />

Michael Esposito<br />

President and<br />

Chief Executive Officer<br />

Louis A. Shapiro<br />

Surgeon-In-Chief and<br />

Medical Director<br />

Todd J. Albert, MD<br />

Executive Vice President and<br />

Chief Operating Officer<br />

Lisa A. Goldstein<br />

Executive Vice President and<br />

Chief Financial Officer<br />

Stacey L. Malakoff<br />

Executive Vice President,<br />

Chief Legal Officer and Secretary<br />

Irene Koch, Esq.<br />

Chairmen Emeriti<br />

Winfield P. Jones<br />

Richard L. Menschel<br />

Aldo Papone<br />

Kendrick R. Wilson III<br />

Credits<br />

Executive Editorial Board<br />

Todd Albert, MD<br />

Mary K. Crow, MD<br />

John Englehart<br />

Lionel Ivashkiv, MD<br />

Louis A. Shapiro<br />

Executive Editor<br />

Rachel Sheehan, MA, MBA<br />

Editor-in-Chief<br />

Deborah Pike Olsen<br />

Writers<br />

Stacey Colino<br />

Beth Howard<br />

Hallie Levine<br />

Design<br />

Suka Creative<br />

Photo Editor<br />

Deborah Garcia<br />

Printing<br />

Earth Color<br />

Photography<br />

John Abbott<br />

Robert Essel


HOSPITAL FOR SPECIAL SURGERY<br />

535 East 70th Street<br />

New York, NY 10021<br />

212-606-1000<br />

hss.edu

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!