1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52

REVIEW ARTICLE Q1

Q16

Tumor necrosis factor inhibitors: clinical utility in autoimmune diseases MARIA A. V. WILLRICH, DAVID L. MURRAY, and MELLISA R. SNYDER ROCHESTER, MN

Tumor necrosis factor (TNF) production is amplified in several autoimmune disorders. In the 1990s, it became a validated therapeutic target used for the treatment of conditions such as rheumatoid arthritis and inflammatory bowel disease. Biologic drugs targeting TNF include engineered monoclonal antibodies and fusion proteins. Currently, there are 5 Food and Drug Administration–approved TNF inhibitors (infliximab, etanercept, adalimumab, certolizumab, and golimumab), representing close to $20 billion in sales. Clinical trials remain open to test their efficacy and safety compared with one another, as well as to assess for clinical outcomes in different conditions and patient populations. The industry is also eager to develop biotherapeutics that are similar but cheaper than the currently existing biologics or are safer with higher efficacy; these are the so-called ‘‘biosimilars.’’ Clinical utility of TNF inhibitors and indications of mono- or combined therapy with immunomodulators are reviewed here. Pharmacokinetics of the TNF inhibitors is affected by routes of administration, clearance mechanisms of immunoglobulins, and immunogenicity. Finally, strategies for management of treatment efficacy and increasing evidence for monitoring of serum concentration of TNF inhibitors are discussed, assessing for the presence of the antidrug antibodies and the different analytical methods available for laboratory testing. As clinical applications of the TNF inhibitors expand, and other classes join the revolution in the treatment of chronic inflammatory disorders, therapeutic drug monitoring of biologics will become increasingly important, with the potential to dramatically improve patient care and management. (Translational Research 2014;-:1–13) Abbreviations: --- ¼ ---

Q3

INTRODUCTION History of therapeutic tumor necrosis factor inhibitors’ development. During the last 2 decades, the chime-

rization and humanization of monoclonal antibodies (mAbs) have led to an exciting new class of biologic

drugs. This class uses the central role that antibodies (aka immunoglobulins [Igs]) play in the adaptive immune system in recognizing and neutralizing antigens. As a class of drugs, the mAbs are engineered to recognize, inhibit, and/or remove human proteins involved in Q4

From the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minn.

1931-5244/$ - see front matter

Submitted for publication May 27, 2014; revision submitted September 9, 2014; accepted for publication September 10, 2014.

http://dx.doi.org/10.1016/j.trsl.2014.09.006

Ó 2014 Elsevier Inc. All rights reserved.

Reprint requests: Mellisa R. Snyder, Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905; e-mail: [email protected].

1 REV 5.2.0 DTD  TRSL830_proof  7 October 2014  6:35 pm  ce

53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93 94 95 96 97 98 99 100 101 102 103 104 105 106

2

107 108 109 110 111 112 113 114 115 116 117 118 119 120 121 122 123 124 125 126 127 128 129 130 131 132 133 134 135 136 137 138 139 140 141 142 143 144 145 146 147 148 149 150 151 152 153 154 155 156 157 158 159 160

Q5

Translational Research - 2014

Willrich et al

disease processes. Tumor necrosis factor a (TNF-a) is one such example. TNF is a cytokine with both proinflammatory and immunoregulatory functions. The role for TNF in rheumatoid arthritis (RA) was identified in 1991.1 Soon after, clinical trials using a chimeric murine–human mAb directed against TNF showed significant clinical efficacy,2,3 validating TNF as a therapeutic target. Since this time, the anti-TNF biologics have revolutionized the treatment of diseases such as RA, ankylosing spondylitis (AS), inflammatory bowel disease (IBD), and psoriasis.4 mAb structure and nomenclature. The basic structure of therapeutic mAbs, such as the TNF inhibitors, is based on the general structure of Igs. Ig molecules are constructed of 4 polypeptide chains including 2 identical heavy chains and 2 identical light chains. Functionally, each antibody can be divided into 2 domains: the variable region (Fab) and the constant region (Fc).5 The Fab fragment contains the complementarity determining regions, which are specifically responsible for antigen binding. The Fc portion of the antibody determines the isotype of the Ig and is identified by the heavy chain (gamma, mu, alpha, delta, epsilon) and the light chain (kappa, lambda). The IgG isotypes in particular have effector functions attractive for therapeutics designed to clear harmful molecules, including complement activation and antibody-dependent cellular toxicity, which is mediated through interaction with specific Fc receptors (FcR). Clearance of IgGs occurs via proteolytic catabolism after receptor-mediated endocytosis in the reticuloendothelial system, resulting from interaction between the antibody and Fc gamma receptors I, II, or III. A second pathway functions to recycle IgGs, which requires binding to the Brambell receptor (FcRn). It is the balance between the proteolytic and FcRn pathways that determines IgG half-life.4 To successfully design a therapeutic mAb, the Ig must be able recognize the human antigen (a function not typically desired in a normal immune response) and still be able to interact with the human FcRn receptors. Therapeutic mAbs have a standardized nomenclature based on their structure. Murine antibodies are named omab (eg, capromab). They were the first in clinical studies; however, their high immunogenic potential, low affinity for FcRn receptors and short half-life compared with a human IgG were major drawbacks. Then, chimeric mAbs (minimum of 65% human) were developed, with murine variable regions and human Fc fractions; those are named ximab (eg, infliximab). Further improvements in therapeutic mAbs came with constructs containing murine hypervariable regions grafted into a human IgG structure, forming humanized mAbs (minimum of 95% human) with the designation

zumab (eg, eculizumab). Lastly, fully engineered human antibodies have been developed, with the naming structure umab (eg, adalimumab and golimumab), that have similar properties to a human IgG and significantly reduced immunogenicity. 5 TNF inhibitors. There are currently 5 Food and Drug Administration (FDA)-approved TNF inhibitors available in the United States (Fig 1). Infliximab, the first biologic in its class, was approved in August 1998 for the treatment of severe Crohn’s disease (CD). In 2002, it was also approved for treatment of refractory RA. Infliximab is a chimeric IgG1 mAb composed of a murine Fab region linked to a human IgG1 kappa constant region produced in cultured Chinese hamster ovary cells. Adalimumab is also produced in Chinese hamster ovary cells; however, it is composed of phagedisplay engineered human-derived variable regions. Golimumab, the newest TNF inhibitor to enter the market, is a human IgG1 kappa, produced in multiple glycoforms by a murine hybridoma cell line.6,7 In contrast to the full-length mAbs, certolizumab is a humanized IgG4 Fab fragment produced by cell culture in Escherichia coli and then chemically linked to polyethylene glycol. This modification of the Fab fragment decreases metabolic clearance rates and extends the half-life of the molecule. Lastly, the most unique of the approved TNF inhibitors is etanercept. Etanercept is a recombinant fusion protein between the TNF p75 receptor and the Fc fraction of a human IgG1. It is currently the most prescribed TNF inhibitor in the US; however, it is not approved for IBD. Since the early 1990s, numerous clinical trials have taken place to address the safety and efficacy of TNF inhibitors in multiple inflammatory conditions and to compare the use of one matched with the other. A total of 92 open clinical trials were identified by searching the term ‘‘TNF inhibitor’’ at http://www.clinicaltrials. gov as of May 13, 2014. The purpose of most of these trials is to evaluate the safety and efficacy of other biologics, such as rituximab (anti-CD20 mAb), vedolizumab (anti-a4b7 integrin mAb), tofacitinib (small molecule inhibitor of JAK 3 kinase), and clazakizumab (anti-IL6 mAb), in patients who are refractory or intolerant to TNF inhibitors. These trials are also addressing outcomes in TNF inhibitor ‘‘switchers,’’ or those patients who fail treatment with one biologic in the class and then move to another one. One new TNF inhibitor, named ozoralizumab, has recently completed phase II clinical trials in patients with RA; however, study results are not yet published. This humanized mAb fragment of approximately 38 KDa is a trivalent, bispecific single-domain antibody against TNF.8 Biosimilars. By 2015, several biologics will lose patent protection in Europe, including infliximab and

REV 5.2.0 DTD  TRSL830_proof  7 October 2014  6:35 pm  ce

161 162 163 164 165 166 167 168 169 170 171 172 173 174 175 176 177 178 179 180 181 182 183 184 185 186 187 188 189 190 191 192 193 194 195 196 197 198 199 200 201 202 203 204 205 206 207 208 209 210 211 212 213 214

Translational Research Volume -, Number -

3

print & web 4C=FPO

215 216 217 218 219 220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235 236 237 238 239 240 241 242 243 244 245 246 247 248 249 250 251 252 253 254 255 256 257 258 259 260 261 262 263 264 265 266 267 268

Willrich et al

Fig 1. TNF inhibitors currently available in the United States. Infliximab is a chimeric antibody with murine amino acid sequences in the Fab region. Adalimumab and golimumab have human amino acid sequences. Certolizumab pegol is a Fab humanized fragment chemically linked to polyethylene glycol. Etanercept is a fusion protein between the TNF receptor extracellular domain and the Fc portion of a human IgG1. It binds not only to TNF, but also to members of the lymphotoxin family, including the soluble LTa3 (figure reproduced with permission from data in Taylor, 20104). Ig, immunoglobulin; TNF, tumor necrosis factor.

etanercept, which has spurred the development of biosimilars.9 According to the World Health Organization, a biosimilar is a biotherapeutic product, which is similar to an already licensed reference product in terms of quality, safety, and efficacy. Similarity is defined as the absence of a relevant difference in the parameter of interest.10 In May 2014, the US FDA released a guideline statement describing the clinical pharmacology data needed to support a biosimilar application.11 Quality is a very important parameter for approval of any drug by the FDA. Although a biosimilar may have an identical amino acid sequence as the reference biologic, heterogeneity of mAbs may happen because of several types of modifications, such as incomplete disulfide bond formation, glycosylation, and oxidation, as well as noncovalent associations with other molecules, conformational diversity, and aggregation. Thousands of variations with the same sequence may coexist.12 Because proof of concept as well as in vitro analytical and in vivo animal studies to the original biotherapeutic are

Q14

needed to account for the molecules variation, most likely the biosimilars market will be in the domain of large pharmaceutical companies, as small biotech companies may not have the necessary resources. A financial analysis has estimated that by 2018 biosimilars will have close to 40% of market share for mAbs in Europe.13 A biosimilar to infliximab, CT-P13, with an identical amino acid sequence and produced in the same type of cell line, has been under development since 1999. Comparative studies for rheumatic diseases have been performed; however, because of regulatory and intellectual property issues, CT-P13 is not currently marketed in the United States, as infliximab’s patent will expire only in September 2018.13 Nonetheless, 2 pharmaceutical companies (Celltrion Biotechnology and Hospira) are filing an application with the FDA in a joint effort, for CT-P13 as the first biosimilar to infliximab (Remsima/ Inflectra). This biosimilar was approved in Europe in Q6 October 2013; FDA approval in the United States is expected in early 2015. Many other biosimilars are in the

REV 5.2.0 DTD  TRSL830_proof  7 October 2014  6:35 pm  ce

269 270 271 272 273 274 275 276 277 278 279 280 281 282 283 284 285 286 287 288 289 290 291 292 293 294 295 296 297 298 299 300 301 302 303 304 305 306 307 308 309 310 311 312 313 314 315 316 317 318 319 320 321 322

4

323 324 325 326 327 328 329 330 331 332 333 334 335 336 337 338 339 340 341 342 343 344 345 346 347 348 349 350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372 373 374 375 376

Translational Research - 2014

Willrich et al

pipeline, according to the Generics and Biosimilars initiative website (http://www.gabionline.net), with some of the main targets being the TNF inhibitors infliximab, adalimumab, and etanercept. CLINICAL UTILITY General introduction. More than 2 million patients have received treatment with at least one of TNF inhibitors since they were approved in 1998 for systemic rheumatic disease and IBD. Over one million of these prescriptions have been for patients with RA.14,15 TNF inhibitors are approved for treatment of various inflammatory conditions within the main categories of systemic rheumatic disease and IBD (Table I). At our institution, for 2013, infliximab was the most frequently billed TNF inhibitor comprising 33% of all TNF biologics, followed closely by adalimumab at 29% and etanercept at 26%. The 2 newest medications, certolizumab and golimumab, were billed at only 10% and 2%, respectively (internal communication). The treatment approach for most chronic inflammatory diseases begins with an induction phase, the goal of which is to induce disease remission, followed by a maintenance phase, designed to maintain remission and prevent disease flares. FDA approval of the TNF inhibitors has addressed their role in both the induction and maintenance phases of treatment.

SYSTEMIC RHEUMATIC DISEASES

Q7

Rheumatoid arthritis. RA is an inflammatory disease of unknown origin, which manifests as an ongoing peripheral polyarthritis. This disabling condition is thought to affect 0.3%–1.2% of the worldwide population.16 If left untreated the disease can lead to debilitating joint destruction through the erosion of cartilage and bone. Being incurable, the goal of treatment is to achieve the lowest possible level of arthritis disease activity, minimizing joint damage, and enhancing physical function. Pharmaceutical interventions are generally classified into 3 broad categories: nonsteroidal antiinflammatory drugs (NSAIDs), corticosteroids, and disease-modifying antirheumatic drugs (DMARDs). The NSAIDS are for the control of inflammation and pain, whereas the corticosteroids and DMARDs are directed toward inhibiting the cellular immune response responsible for the inflammation. The goal of any treatment is to place the patient into strict complete remission. However, strict remissions by the preliminary criteria of the American College of Rheumatology (ACR)17 are rare and usually transient.18,19 Clinical outcomes can be improved using treatment strategies with predefined targets and tight control of disease activity.20,21 In light of these

findings, a 2010 international task force emphasized that the goal of treatment in RA should be remission.20 TNF inhibitors were the first of the biological DMARDs to be approved for the treatment of RA and, over the past decade, have become part of the routine treatment of patients with this disease. All 5 of the current TNF inhibitors are FDA approved for the treatment of RA. ACR treatment recommendations for this class of drugs are based on the duration of disease activity (greater or less than 6 months), disease activity (low, moderate, and high), and clinical features which correspond to prognosis. With the exception of early high disease activity with poor clinical prognosis, the use of TNF inhibitors is indicated only after patients have failed other therapies.22 In clinical trials, the combination of methotrexate and anti-TNF therapy is the most efficacious in showing reductions evidenced by radiographic damage.14 Juvenile RA. Juvenile RA is not a single disease, but is a term that encompasses all forms of arthritis, which have persisted for more than 6 weeks in patients younger than 16 years. It is classified in different subsets based on associated symptoms and number of joints involved. TNF inhibitors are used more commonly for the polyarthritic manifestations of disease, where they have proven efficacy.23-25 For systemic juvenile RA, TNF inhibitors have shown little efficacy.26 Etanercept is approved in patients aged 2 years or older as a monotherapy. Adalimumab is approved in pediatric patients aged 4 years and older, being used alone or with methotrexate. Ankylosing spondylitis. AS is a form of spondyloarthritis with great disease burden. It is a chronic inflammatory disease affecting the axial skeleton resulting in back pain and progressive stiffness of the spine. AS typically develops in young adults, between 20 and 30 years of age. It may also involve hips, shoulders, and peripheral joints. There is a strong correlation between the presence of the human leukocyte antigen B27 and incidence of AS. The prevalence of AS is estimated to vary between 0.1% and 1%.27 Multiple disease activity scores are available and have become common ways of managing the condition. Standard treatment options, including NSAIDs and opioids, are seldom effective during active AS. Infliximab was the first TNF inhibitor to show efficacy in AS clinical trials, improving signs, symptoms, function, and quality of life 2 weeks after the initiation of therapy.28 Adalimumab, golimumab, and etanercept have also shown similar efficacy.29-31 The combination of a TNF inhibitor with methotrexate does not seem to improve outcomes in AS.32,33 Adult plaque psoriasis and psoriatic arthritis. Plaque psoriasis (PsO) is a disease of young adults who present with symmetrically distributed demarcated

REV 5.2.0 DTD  TRSL830_proof  7 October 2014  6:35 pm  ce

377 378 379 380 381 382 383 384 385 386 387 388 389 390 391 392 393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430

431 432 433 434 435 436 437 438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484 Translational Research Volume -, Number -

REV 5.2.0 DTD  TRSL830_proof  7 October 2014  6:35 pm  ce

Table I. Characteristics of the FDA-approved TNF inhibitors Infliximab

Adalimumab

Origin

Chimeric, murine Fab

Biologic type Molecular weight (kDa) Mode of production

IgG1 kappa 150 Culture in CHO cells

Fully human, recombinant human Fab IgG1 kappa 150 Fab produced by phage display, cell culture in CHO cells Subcutaneous every other week 10–20 2002

Route of administration

Intravenous every 6–8 weeks Half-life of molecule (days) 8–10 Year of first FDA approval 1998 Conditions where treatment is FDA approved Rheumatoid arthritis X Psoriatic arthritis X Ankylosing spondylitis X Adult plaque psoriasis X Juvenile rheumatoid arthritis Adult Crohn’s disease X Pediatric Crohn’s disease X Adult ulcerative colitis X Pediatric ulcerative colitis X

Golimumab

Certolizumab

Etanercept

Fully human

Humanized Fab

TNF receptor fusion protein

IgG1 kappa 150 Produced in transgenic mice

Fab of IgG4 91 Expressed in Escherichia coli and conjugated to polyethylene glycol Subcutaneous every other week 14 2008

Fc of IgG1 150 DNA recombinant technology in CHO cells

X

X X X X X

Subcutaneous once a month 14 2009

X X X X X X

X X X

X

X

Q2

Subcutaneous once or twice a week 4 1998

X

Abbreviations: CHO, Chinese hamster ovary; FDA, Food and Drug Administration; TNF, tumor necrosis factor.

Willrich et al 5

485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538

6

539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592

Translational Research - 2014

Willrich et al

erythematous plaques, involving the scalp, elbows, knees, and back. There is increasing evidence supporting the recognition of psoriasis as a multisystem chronic inflammatory disorder, with multiple associated comorbidities, including extracutaneous disorders such as psoriatic arthritis (PsA). PsA generally manifests as an inflammatory arthritis associated with psoriasis, which can significantly affect a patient’s quality of life. Pain and stiffness in affected joints are common symptoms, with significant joint deformity.34 NSAIDs, physical therapy, and DMARDs were the only treatment options for these conditions before TNF inhibitors became available. For PsO, topical therapies and ultraviolet light are recommended first. Systemic treatment options include DMARDs and infliximab, adalimumab, and etanercept as TNF inhibitors. For moderate to severely active PsA, infliximab, adalimumab, golimumab, and etanercept are considered first line of treatment,35-37 as they have shown marked improvements in these conditions. Inflammatory bowel disease. IBD consists of 2 unique diseases—ulcerative colitis (UC) and CD. Both UC and CD are chronic diseases resulting from inflammation within the gastrointestinal tract.38,39 The inflammation appears to be initiated by an uncontrolled immune response, presumably against flora present within the gut. It is estimated that nearly one million people in the United States are affected with IBD. UC and CD can be distinguished from one another based on clinical symptoms, affected sites within the gastrointestinal tract, and patterns of inflammation. UC tends to be restricted to the colon and is characterized by continuous inflammation of the mucosa. In contrast, CD may involve any area of the gastrointestinal tract, with inflammation that tends to be patchy and transmural, sometimes with evidence of granulomas. Standard therapy differs significantly between CD and UC, and is also affected by the site and extent of inflammation and disease severity. In general, first-line treatment for IBD centers on immunosuppressive therapy, such as corticosteroids, azathioprine, or methotrexate. Antibiotics have also proven to be effective for some patients.40,41 Infliximab and adalimumab are both approved for the treatment of moderate to severe CD and UC in adult patients who have demonstrated inadequate responses to standard therapies, such as antibiotics, corticosteroids, and aminosalicylates. Both are approved for induction and maintenance phases of treatment. In contrast, certolizumab is only approved for moderate to severe CD as a second-line therapy in adults, whereas golimumab is approved only for moderate to severe UC in adult patients who have failed previous therapies or who are steroid dependent. For treatment of pediatric IBD, infliximab is the only TNF inhibitor that

is approved by the FDA, specifically for children aged .6 years.42 Potential adverse effects. Adverse effects associated with TNF inhibitor therapy have been identified both through clinical trials and postmarket surveillance programs, such as the FDA Adverse Event Reporting System. Some adverse effects are relatively mild and can be managed such that discontinuation of the TNF blocking treatment is not necessary. However, other adverse effects are more serious requiring patients to change to another class of agents for treatment of their inflammatory disease (Fig 2). Infusion reactions, both acute and delayed, have been documented, particularly with infliximab. Acute reactions usually occur within 4 hours of infusion. Although sometimes associated with production of IgE antibodies, most acute reactions are actually anaphylactoid, or nonallergic, in nature. Treatment of acute reactions generally involves stopping the infusion, at least temporarily, and administration of diphenhydramine. Delayed infusion reactions, in contrast, appear to be a type III hypersensitivity reaction, mediated by production of immune complexes. Symptoms of a delayed reaction include rash, arthralgias, myalgias, and fatigue. Delayed infusion reactions seem to be related to the immunogenicity of the TNF inhibitor and the presence of antidrug antibodies. Because of this, strategies to reduce the immune response against the biologic, such as administration of methotrexate or azathioprine, can be helpful in preventing delayed infusion reactions.43,44 Because TNF plays an important proinflammatory role in the immune response, it should not be surprising that blockage of this pathway leads to increases in rates of infection. A variety of infections have been evaluated in the context of TNF inhibitor therapy, including mycobacterial, bacterial, fungal, and viral. Treatment with TNF inhibitors is associated with an increased incidence of mycobacterial tuberculosis (TB) infections, mostly attributed to reactivation of latent infection. Data from the FDA Adverse Event Reporting System have estimated that the incidences of TB infections were 54 and 28 per 100,000 patient years on infliximab and etanercept, respectively, which is 4- to 10-fold higher than in the general population. Bacterial infections also appear to be an issue.45 A study in hospitalized patients with RA has demonstrated that treatment with a TNF inhibitor resulted in an odds ratio of 4.2 for development of a bacterial infection within 6 months of initiation of Q8 treatment.45,46 Similar to TB, it appears that the risk of bacterial infection is greater with infliximab compared with etanercept. TNF inhibitors also appear to be a risk factor for fungal infection, likely because of impairments in granuloma formation. In 2008, the FDA released a statement warning of the risk of

REV 5.2.0 DTD  TRSL830_proof  7 October 2014  6:35 pm  ce

593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 643 644 645 646

Translational Research Volume -, Number -

7

web 4C=FPO

647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668 669 670 671 672 673 674 675 676 677 678 679 680 681 682 683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700

Willrich et al

Fig 2. Main adverse effects of the use of TNF inhibitors. DMARDs, disease-modifying antirheumatic drugs; TNF, tumor necrosis factor.

REV 5.2.0 DTD  TRSL830_proof  7 October 2014  6:35 pm  ce

Q15

701 702 703 704 705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720 721 722 723 724 725 726 727 728 729 730 731 732 733 734 735 736 737 738 739 740 741 742 743 744 745 746 747 748 749 750 751 752 753 754

8

755 756 757 758 759 760 761 762 763 764 765 766 767 768 769 770 771 772 773 774 775 776 777 778 779 780 781 782 783 784 785 786 787 788 789 790 791 792 793 794 795 796 797 798 799 800 801 802 803 804 805 806 807 808

Q9

Translational Research - 2014

Willrich et al

invasive fungal infections for patients receiving a TNF inhibitor. Lastly, the risk of viral infections for patients on TNF therapy is more of a question. There have been conflicting reports in the literature, which may be because of different effects associated with various viruses. On the basis of all available data, it is recommended that TNF inhibitor therapy be avoided in patients with any active infection, including bacterial and viral.47-55 In addition, all patients should be screened for TB, hepatitis B, hepatitis C, and possibly also HIV infections, and appropriately treated before initiating TNF inhibitor therapy. One of the original properties characterized for TNF was its ability to lyse tumor cells. This raises the question of whether blocking this pathway could lead to an increased risk of malignancy. The first evidence of this potential association was an increased risk of lymphomas, primarily non-Hodgkins, in patients on infliximab or etanercept therapy. Over the years, numerous studies have addressed this issue, with mixed results.50 As a result, whether TNF inhibitor therapy results in a general increased risk of malignancy, whether it is specific for certain biologics or disease states, or whether it is restricted to patients with an underlying predisposition to cancer is unclear. However, in 2004, a statement was added to the infliximab label, warning of the lymphomas risk. Hopefully, continue surveillance and prospective studies will help us to address this potentially significant adverse effect. Reports of neurologic disorders have also been associated with TNF inhibitors. In a case of aseptic meningitis in a 53-year-old man receiving infliximab for RA, the abnormalities in the cerebrospinal fluid had cleared on repeat examination a month later, which suggested a transient drug reaction rather than a chronic inflammatory neurologic disorder.56 Infliximab has also been associated with a worsening of multiple sclerosis in 2 patients,57 and at least 4 cases of demyelination with etanercept, which all had resolved by 6 weeks, have been reported to the US FDA. Therefore, TNF inhibitors should not be given to people with a history of demyelinating disease, and they should be withdrawn if such a disorder develops. Pharmacokinetics. Although the TNF inhibitors and therapeutic mAbs have been available for more than 2 decades, knowledge of the factors that affect pharmacokinetics is still poorly elucidated. A greater understanding of how mAb therapeutics are metabolized is of critical importance, as studies have shown, in a variety of disease states, that circulating concentrations of the drug are linked to clinical efficacy. There are certain patient-specific characteristics that impact the pharmacokinetics of the therapeutic TNF mAbs. Particularly relevant to chronic autoimmune

diseases is that systemic inflammation affects mAb clearance. Increased C-reactive protein (CRP), increased TNF-a, and hypoalbuminemia are all associated with decreased concentrations of TNF inhibitors. In addition, hypergammaglobulinemia also may result in accelerated clearance of the mAb, which is likely attributable to saturation of the FcRn recycling system by endogenous Igs. It also appears, at least for infliximab, that body mass and male gender are risk factors for decreased circulating concentrations.58 One exogenous parameter that affects pharmacokinetics is the route of administration. Infliximab is administered intravenously, whereas all other TNF inhibitors are self-administered with subcutaneous injections. Large volumes of drug may be delivered intravenously, compared with the relatively small volumes that can be administered subcutaneously. In addition, the differences between peak and trough concentrations of the drug are much larger when given by intravenous (IV) infusion, in comparison with the more consistent concentrations observed with subcutaneous delivery. This is likely because of the fact that IV administration has a more rapid distribution, which is contrasted by the slower absorption after subcutaneous injections. Also, with IV administration, mAbs are more readily bioavailable with less interindividual variability and reduced immunogenicity.58 With subcutaneous injections, absorption likely occurs through lymphatic drainage, where antigen processing increases the immunogenic properties of the mAb.58 All mAb therapeutics, including the TNF inhibitors, are ‘‘foreign’’ proteins, with the potential to be recognized by the adaptive immune system. The presence of the therapeutic mAb may activate a humoral immune response, resulting in the production of antidrug antibodies. This property is referred to as immunogenicity. The presence of antidrug antibodies has a substantial effect on the pharmacokinetics of the TNF inhibitors. Most studies have demonstrated that the presence of antidrug antibodies is associated with significantly lower concentrations of circulating therapeutic mAb. This likely results from rapid phagocytic clearance of immune complexes that form between the antidrug antibody and the therapeutic mAb. This subsequently leads to decreased concentration of the TNF inhibitor in circulation. The immunogenic potential of the TNF inhibitor is closely linked to the degree of humanization of the mAb. Incidence of antibodies against the human/ murine chimeric infliximab has been shown to be as Q10 high as 12%–70% of patients receiving the drug.59 However, it has also been shown that adalimumab, which is more fully humanized, is also immunogenic.60,61 Etanercept has what appears to be the lowest immunogenic potential, being composed only of the

REV 5.2.0 DTD  TRSL830_proof  7 October 2014  6:35 pm  ce

809 810 811 812 813 814 815 816 817 818 819 820 821 822 823 824 825 826 827 828 829 830 831 832 833 834 835 836 837 838 839 840 841 842 843 844 845 846 847 848 849 850 851 852 853 854 855 856 857 858 859 860 861 862

Translational Research Volume -, Number -

863 864 865 866 867 868 869 870 871 872 873 874 875 876 877 878 879 880 881 882 883 884 885 886 887 888 889 890 891 892 893 894 895 896 897 898 899 900 901 902 903 904 905 906 907 908 909 910 911 912 913 914 915 916

Willrich et al

TNF receptor extracellular domain and human Ig sequences. In fact, the incidence of anti-etanercept antibodies appears to be relatively low, in comparison with infliximab and adalimumab. The humanized anti-TNF inhibitors certolizumab and golimumab likely share similar immunogenic properties to adalimumab. However, studies assessing for the incidence of anticertolizumab and anti-golimumab antibodies are lacking in the public literature. The link between antidrug antibodies, decreased drug concentration, and clinical efficacy is clear. As such, strategies aimed at reducing the incidence antidrug antibodies can lead to improved outcomes for patients receiving a TNF inhibitor. Two approaches demonstrated to reduce the immunogenicity of the TNF mAbs are coadministration of an immunomodulator and scheduled maintenance dosing. Combination therapy of infliximab or adalimumab and an immunosuppressive agent has been demonstrated to decrease the incidence of antidrug antibodies in comparison with the TNF inhibitor alone. There does not appear to be significant differences between methotrexate and azathioprine, which are the most commonly used immunosuppressive agents for combination therapy.14,62 Some studies have further shown that the decreased incidence of antidrug antibodies in combination therapy is associated with increased concentrations of the TNF inhibitor and improved clinical outcomes.59,63,64 However, there is some concern that this approach may lead to overimmunosuppression and an increased risk of infection and malignancy. This may especially be a concern for patients on long-term maintenance therapy. In addition to combination therapy, scheduled treatment as opposed to episodic treatment appears to offer some advantages related to the formation of antidrug antibodies. Scheduled treatment is defined as a standardized approach to maintenance therapy, rather than episodic treatment driven by disease flares or worsening clinical symptoms. Particularly with infliximab, decreased incidence of antidrug antibodies and improved clinical outcomes have been seen in patients on scheduled rather than episodic maintenance therapy.32 It appears that a combination of the 2 strategies, scheduled therapy of anti-TNF, and a concomitant immunosuppressant, may alleviate some of the immunogenicity of these biological agents. Management and monitoring of TNF inhibitors. For RA, monitoring of disease activity is achieved using disease activity scores. The most commonly used is a Simplified Disease Activity Index, comprising evaluation of tender and swollen joint counts, patient and physician global assessments, and CRP concentrations. The index is used by the ACR and European League Against Rheumatism for the definition of remission.65 Patients are

9

usually evaluated at least every 3 months to monitor response to therapy using the same measures, and adjustments to therapy are made to maintain control of disease activity if targeted treatment goals (remission or low disease activity) are not achieved. Response criteria are defined for both moderate and major changes in disease activity and are applied to patients in clinical trials. A common response criterion is defined by the ACR, in which the variables outline a 20% improvement (ACR20). Given the relatively high cost of TNF inhibitor treatment, several health systems have set up monitoring programs to track usage and outcomes. The Swedish biologics register ‘‘Anti-Rheumatic Therapy in Sweden,’’ a nationwide database on patients prescribed biologic therapies for the treatment of rheumatic diseases, has been active since biologics were first available in clinical practice in 1999. The cohort in this registry is quite large covering 87% of the RA patients.66 Within this cohort, the TNF inhibitors represent the vast majority of prescribed biologics. Analysis of this cohort showed half of patients starting infliximab, adalimumab, or etanercept during the period 2005–2012 discontinued treatment for various reasons. The most common reason for discontinuing treatment was nonresponse. Of these patients, one-third switched within 2 months to a second TNF inhibitor (infliximab, etanercept, or adalimumab). Around 35% of patients achieved low disease activity or remission at 6 months. Best results were observed among patients who switched from infliximab to etanercept because of (secondary) inefficacy. Etanercept as a second TNF inhibitor was associated with longer drug survival compared with infliximab.67 A similar national drug registry, BIOBADASER 2.0, is composed of pa- Q11 tients with rheumatic diseases treated with any biological agent in 14 large public university hospitals in Spain.68 Patients entering the registry are followed prospectively and evaluated at the time when an adverse event or a change in the biological occurs. A total of 2907 RA patients were included in the study. This group found similar results to the Anti-Rheumatic Therapy in Sweden cohort. Patients receiving their first TNF inhibitor were discontinued in the first year at a rate of 17%, 19%, and 21% in 2000–2003, 2004–2006, and 2007– 2009, respectively (no statistical differences between the drugs, P 5 0.074). In the early years, the main reason for discontinuation of a TNF inhibitor was adverse reactions, whereas in recent years the most common reason for discontinuation was inefficacy. Given the relatively high rate of loss of efficacy, some studies have been undertaken to measure the drug concentrations in patients treated with TNF antagonists. Jamnitski et al69 measured drug levels in 292 RA patients on etanercept and demonstrated that lower drug

REV 5.2.0 DTD  TRSL830_proof  7 October 2014  6:35 pm  ce

917 918 919 920 921 922 923 924 925 926 927 928 929 930 931 932 933 934 935 936 937 938 939 940 941 942 943 944 945 946 947 948 949 950 951 952 953 954 955 956 957 958 959 960 961 962 963 964 965 966 967 968 969 970

10

971 972 973 974 975 976 977 978 979 980 981 982 983 984 985 986 987 988 989 990 991 992 993 994 995 996 997 998 999 1000 1001 1002 1003 1004 1005 1006 1007 1008 1009 1010 1011 1012 1013 1014 1015 1016 1017 1018 1019 1020 1021 1022 1023 1024

Translational Research - 2014

Willrich et al

levels were associated with nonresponse. A similar study was performed by Daien et al,70 which had recommended that etanercept concentrations measured 3 months post-treatment could explain the absence of response. However, there are currently no recommendations for following drug concentrations in RA patients as part of routine clinical care. Loss of response is also associated with the development of antidrug antibodies. However, only a limited number of studies have been done on etanercept and infliximab in patients with RA. In other rheumatic diseases, such as AS, PsA, and PsO, no guidance on whether monitoring TNF inhibitors is associated with improved outcomes is available. In IBD, multiple systems have been validated for assessment of disease activity. Most of these systems are specifically designed for either CD or UC. For CD, the Clinical Disease Activity Index is the most commonly used, and includes clinical variables (number of liquid stools per day, abdominal pain, and number of complications), patient characteristics (body weight), and laboratory criteria (hematocrit). This scoring system can be used to assess for response, induction of remission, and maintenance of remission based on the absolute score or a change in score compared with baseline value. There are also several tools for evaluation of endoscopic response to treatment. These systems are based on assessment of various types of lesions evident on endoscopy. Similarly, various systems have been developed for use in UC. Many of these systems combine clinical evaluation, laboratory testing, and endoscopic findings into a single score. For IBD, there is accumulating evidence that the circulating concentrations of the TNF inhibitor and the presence of antidrug antibodies are associated with clinical outcomes. Low trough concentrations of infliximab, for instance, are associated with a shorter duration of response and less mucosal healing, and are inversely correlated with clinical and endoscopic remission in CD.62,71 The same holds true for certolizumab, where higher concentrations of the drug were associated with endoscopic response and remission in a clinical trial with 89 adult patients with moderate to severe CD, whereas lower certolizumab concentrations were associated with increased CRP and greater body weight.72 Because of the association with clinical responses, assessment of TNF inhibitor concentrations and antidrug antibodies is increasingly being advocated for use in routine clinical practice in patients with IBD.38,73-75 According to a recent study, the primary indications for testing of infliximab concentrations in the context of IBD were loss of response, partial response on initiation of therapy, and autoimmune or

hypersensitivity reactions.76 Specifically in the context of loss of response or lack of initial response, assessment of drug concentration and antidrug antibodies has the potential to directly impact patient management. For example, patients with decreased concentrations of the TNF inhibitor but no detectable antidrug antibodies may benefit from dose escalation. In contrast, this approach is less likely to be effective in patients with decreased concentrations of drug in the presence of antidrug antibodies. For these individuals, a switch to another TNF inhibitor may be indicated. In some cases, patients will be found to have an apparently adequate concentration of the TNF inhibitor. This may occur in the presence or absence of antidrug antibodies. Regardless, loss of response in the context of adequate concentrations of drug may suggest that the patient is not responding to TNF inhibition and that perhaps another class of biologics may be beneficial.38,62,72,74,75,77,78 However, this raises the question of what is the optimal therapeutic dose for the various TNF inhibitors. Some studies have addressed this for infliximab and adalimumab, although there is some variation in the data.61,62,76,79-82 In contrast, there are no data in the literature on therapeutic targets for etanercept, certolizumab, and golimumab. Analytical perspectives. Given the frequency of loss of response to biologics and the presence of antidrug antibodies, methods to measure drug concentrations in patients on TNF inhibitors are useful in patient management.62 As a class of drugs, the therapeutic mAbs present a unique challenge for serum quantitation in comparison with traditional small molecule drugs. For the small molecule drugs, isolation of the analyte from background proteins can be accomplished by precipitation, extraction, or size exclusion chromatography. The mAbs are not only part of the protein background; they are also structurally very similar to the native high abundance Igs. First-generation assays developed for measurement of serum TNF inhibitors and antidrug antibodies were based on radioimmunoassays83 and solid-phase, double-antigen, and enzyme-linked immunosorbent assay.84 An analyte specific method using cell lines that have a TNF sensitive reporter gene has demonstrated to quantitate the activity of TNF in patients treated with TNF inhibitors.85 A liquid-phase mobility shift assay for measurement of TNF inhibitors and antidrug antibodies in the presence of drug has been described for infliximab and adalimumab.61,86 The mobility shift assay is based on the shift in retention time of the antigen-antibody immunocomplex vs free antigen on size-exclusion chromatography. A proteomics-based assay for the quantitation of TNF inhibitors using tryptic peptides and mass spectrometry has also been

REV 5.2.0 DTD  TRSL830_proof  7 October 2014  6:35 pm  ce

1025 1026 1027 1028 1029 1030 1031 1032 1033 1034 1035 1036 1037 1038 1039 1040 1041 1042 1043 1044 1045 1046 1047 1048 1049 1050 1051 1052 1053 1054 1055 1056 1057 1058 1059 1060 1061 1062 1063 1064 1065 1066 1067 1068 1069 1070 1071 1072 1073 1074 1075 1076 1077 1078

Translational Research Volume -, Number -

1079 1080 1081 1082 1083 1084 1085 1086 1087 1088 1089 1090 1091 1092 1093 1094 1095 1096 1097 1098 1099 1100 1101 1102 1103 1104 1105 1106 1107 1108 1109 1110 1111 1112 1113 1114 1115 1116 1117 1118 1119 1120 1121 1122 1123 1124 1125 1126 1127 1128 1129 1130 1131 1132

Willrich et al

described87 and may overcome some of the limitations of immunoassays, with the potential of being more costeffective. CONCLUSIONS

TNF inhibitors are an important class of biologics for the treatment of chronic inflammatory conditions such as RA and IBD. Monitoring for the concentration of the TNF inhibitor and the presence of antidrug antibodies in patients is associated with improved outcomes; however, clinical assays are expensive and not yet widely available, so most presented studies are retrospective. As more affordable and better methods become available to quantitate TNF inhibitors in serum of patients undergoing therapy, it will allow for the execution of robust prospective studies to better understand pharmacokinetics, immunogenicity, and therapeutic thresholds to optimize therapy. As the applications of the TNF inhibitors expand, and other classes of biologics join the revolution in the treatment of chronic inflammatory disorders, complete elucidation of mechanism of action of biologics and therapeutic monitoring with laboratory tests will become increasingly important, saving costs and at the same time guiding providers’ decisions instead of empirically adjusting doses or switching therapies. Moreover, it has the potential to ease the burden of uncontrolled disease and dramatically improve patient care and therapy success rates. ACKNOWLEDGMENTS

Conflicts of Interest: All authors declare no conflict of interest concerning any financial or personal relationship with organizations that could potentially be perceived as influential on the described research. All authors have read and agreed on the journal’s authorship agreement and policy on disclosure of potential conflicts of interest. REFERENCES

1. Keffer J, Probert L, Cazlaris H, et al. Transgenic mice expressing human tumour necrosis factor: a predictive genetic model of arthritis. EMBO J 1991;10:4025–31. 2. Elliott MJ, Maini RN, Feldmann M, et al. Randomised doubleblind comparison of chimeric monoclonal antibody to tumour necrosis factor alpha (cA2) versus placebo in rheumatoid arthritis. Lancet 1994;344:1105–10. 3. Elliott MJ, Maini RN, Feldmann M, et al. Repeated therapy with monoclonal antibody to tumour necrosis factor alpha (cA2) in patients with rheumatoid arthritis. Lancet 1994;344:1125–7. 4. Taylor PC. Pharmacology of TNF blockade in rheumatoid arthritis and other chronic inflammatory diseases. Curr Opin Pharmacol 2010;10:308–15. 5. Abbas AK, Lichtman AH, Pillai S. Antibodies and antigens. In: Abbas AK, ed. Cellular and molecular immunology. 6th ed. Philadelphia: Saunders Elsevier, 2007.

11

6. Hutas G. Golimumab, a fully human monoclonal antibody against TNFalpha. Curr Opin Mol Ther 2008;10:393–406. 7. Mazumdar S, Greenwald D. Golimumab. MAbs 2009;1:422–31. 8. Kratz F, Elsadek B. Clinical impact of serum proteins on drug delivery. J Control Release 2012;161:429–45. 9. Dimitrov DS. Therapeutic proteins. Methods Mol Biol 2012;899: 1–26. 10. Expert Committee on Biological Standardization. Guidelines on evaluation of similar biotherapeutics products (SBPs). Annex 2009;2:977. 11. FDA. Guidance for industry: clinical pharmacology data to support a demonstration of biosimilarity to a reference product. Silver Spring. Available from: http://www.fda.gov/downloads/Drugs/ GuidanceComplianceRegulatoryInformation/Guidances/UCM39 7017.pdf; 2014. Q12 12. Liu H, Gaza-Bulseco G, Faldu D, Chumsae C, Sun J. Heterogeneity of monoclonal antibodies. J Pharm Sci 2008;97:2426–47. 13. Kay J, Smolen JS. Biosimilars to treat inflammatory arthritis: the challenge of proving identity. Ann Rheum Dis 2013;72:1589–93. 14. Taylor PC, Feldmann M. Anti-TNF biologic agents: still the therapy of choice for rheumatoid arthritis. Nat Rev Rheumatol 2009; 5:578–82. 15. Remicade website for Healthcare Professionals. 2014 [cited 2014 May 20, 2014]; Available from: (http://www.remicade.com/hcp) 16. Beresniak A, Ariza-Ariza R, Garcia-Llorente JF, RamirezArellano A, Dupont D. Modelling cost-effectiveness of biologic treatments based on disease activity scores for the management of rheumatoid arthritis in Spain. Int J Inflam 2011;2011:727634. 17. Pinals RS, Masi AT, Larsen RA. Preliminary criteria for clinical remission in rheumatoid arthritis. Arthritis Rheum 1981;24: 1308–15. 18. Makinen H, Kautiainen H, Hannonen P, et al. Disease activity score 28 as an instrument to measure disease activity in patients with early rheumatoid arthritis. J Rheumatol 2007;34:1987–91. 19. Sokka T, Makinen H, Hannonen P, Pincus T. Most people over age 50 in the general population do not meet ACR remission criteria or OMERACT minimal disease activity criteria for rheumatoid arthritis. Rheumatology 2007;46:1020–3. 20. Schoels M, Aletaha D, Smolen JS, et al. Follow-up standards and treatment targets in rheumatoid arthritis: results of a questionnaire at the EULAR 2008. Ann Rheum Dis 2010;69:575–8. 21. Schoels M, Knevel R, Aletaha D, et al. Evidence for treating rheumatoid arthritis to target: results of a systematic literature search. Ann Rheum Dis 2010;69:638–43. 22. Singh JA, Furst DE, Bharat A, et al. 2012 update of the 2008 American College of Rheumatology recommendations for the use of disease-modifying antirheumatic drugs and biologic agents in the treatment of rheumatoid arthritis. Arthritis Care Res 2012; 64:625–39. 23. Lovell DJ, Giannini EH, Reiff A, et al. Etanercept in children with polyarticular juvenile rheumatoid arthritis. Pediatric Rheumatology Collaborative Study Group. N Engl J Med 2000;342:763–9. 24. Lovell DJ, Ruperto N, Goodman S, et al. Adalimumab with or without methotrexate in juvenile rheumatoid arthritis. N Engl J Med 2008;359:810–20. 25. Ruperto N, Lovell DJ, Cuttica R, et al. A randomized, placebocontrolled trial of infliximab plus methotrexate for the treatment of polyarticular-course juvenile rheumatoid arthritis. Arthritis Rheum 2007;56:3096–106. 26. Prakken B, Albani S, Martini A. Juvenile idiopathic arthritis. Lancet 2011;377:2138–49. 27. Dean LE, Jones GT, MacDonald AG, Downham C, Sturrock RD, Macfarlane GJ. Global prevalence of ankylosing spondylitis. Rheumatology (Oxford) 2014;53:650–7.

REV 5.2.0 DTD  TRSL830_proof  7 October 2014  6:35 pm  ce

1133 1134 1135 1136 1137 1138 1139 1140 1141 1142 1143 1144 1145 1146 1147 1148 1149 1150 1151 1152 1153 1154 1155 1156 1157 1158 1159 1160 1161 1162 1163 1164 1165 1166 1167 1168 1169 1170 1171 1172 1173 1174 1175 1176 1177 1178 1179 1180 1181 1182 1183 1184 1185 1186

12

1187 1188 1189 1190 1191 1192 1193 1194 1195 1196 1197 1198 1199 1200 1201 1202 1203 1204 1205 1206 1207 1208 1209 1210 1211 1212 1213 1214 1215 1216 1217 1218 1219 1220 1221 1222 1223 1224 1225 1226 1227 1228 1229 1230 1231 1232 1233 1234 1235 1236 1237 1238 1239 1240

Q13

Translational Research - 2014

Willrich et al

28. Braun J, Landewe R, Hermann KG, et al. Major reduction in spinal inflammation in patients with ankylosing spondylitis after treatment with infliximab: results of a multicenter, randomized, double-blind, placebo-controlled magnetic resonance imaging study. Arthritis Rheum 2006;54:1646–52. 29. Callhoff J, Sieper J, Weiss A, Zink A, Listing J. Efficacy of TNFalpha blockers in patients with ankylosing spondylitis and non-radiographic axial spondyloarthritis: a meta-analysis. Ann Rheum Dis 2014. 30. McLeod C, Bagust A, Boland A, et al. Adalimumab, etanercept and infliximab for the treatment of ankylosing spondylitis: a systematic review and economic evaluation. Health Technol Assess 2007;11:1–158. iii–iv. 31. Sieper J, Rudwaleit M, Braun J. Adalimumab for the treatment of ankylosing spondylitis. Expert Opin Pharmacother 2007;8:831–8. 32. Breban M, Ravaud P, Claudepierre P, et al. Maintenance of infliximab treatment in ankylosing spondylitis: results of a one-year randomized controlled trial comparing systematic versus ondemand treatment. Arthritis Rheum 2008;58:88–97. 33. Mulleman D, Lauferon F, Wendling D, et al. Infliximab in ankylosing spondylitis: alone or in combination with methotrexate? A pharmacokinetic comparative study. Arthritis Res Ther 2011; 13:R82. 34. Oriente P, Biondi-Oriente C, Scarpa R. Psoriatic arthritis. Clinical manifestations. Baillieres Clin Rheumatol 1994;8:277–94. 35. Gossec L, Smolen JS, Gaujoux-Viala C, et al. European League Against Rheumatism recommendations for the management of psoriatic arthritis with pharmacological therapies. Ann Rheum Dis 2012;71:4–12. 36. Papoutsaki M, Costanzo A. Treatment of psoriasis and psoriatic arthritis. BioDrugs 2013;27(Suppl 1):3–12. 37. Ritchlin CT, Kavanaugh A, Gladman DD, et al. Treatment recommendations for psoriatic arthritis. Ann Rheum Dis 2009;68: 1387–94. 38. Eser A, Primas C, Reinisch W. Drug monitoring of biologics in inflammatory bowel disease. Curr Opin Gastroenterol 2013;29:391–6. 39. Mowat C, Cole A, Windsor A, et al. Guidelines for the management of inflammatory bowel disease in adults 2011;60:571–607. 40. Kornbluth A, Sachar DB. Ulcerative colitis practice guidelines in adults: American College of Gastroenterology, Practice Parameters Committee. Am J Gastroenterol 2010;105:501–23. quiz 24. 41. Lichtenstein GR, Hanauer SB, Sandborn WJ. Management of Crohn’s disease in adults. Am J Gastroenterol 2009;104: 465–83. quiz 4, 84. 42. Bernstein CN, Fried M, Krabshuis JH, et al. World Gastroenterology Organization Practice Guidelines for the diagnosis and management of IBD in 2010. Inflamm Bowel Dis 2010;16:112–24. 43. Lichtenstein GR. Comprehensive review: antitumor necrosis factor agents in inflammatory bowel disease and factors implicated in treatment response. Therap Adv Gastroenterol 2013;6:269–93. 44. Steenholdt C, Svenson M, Bendtzen K, Thomsen OO, Brynskov J, Ainsworth MA. Severe infusion reactions to infliximab: aetiology, immunogenicity and risk factors in patients with inflammatory bowel disease. Aliment Pharmacol Ther 2011;34:51–8. 45. Raychaudhuri SP, Nguyen CT, Raychaudhuri SK, Gershwin ME. Incidence and nature of infectious disease in patients treated with anti-TNF agents. Autoimmun Rev 2009;9:67–81. 46. Mohan VP, Scanga CA, Yu K, et al. Effects of tumor necrosis factor alpha on host immune response in chronic persistent tuberculosis: possible role for limiting pathology. Infect Immun 2001;69: 1847–55. 47. Baert F, Noman M, Vermeire S, et al. Influence of immunogenicity on the long-term efficacy of infliximab in Crohn’s disease. N Engl J Med 2003;348:601–8.

48. Murdaca G, Spano F, Puppo F. Selective TNF-alpha inhibitorinduced injection site reactions. Expert Opin Drug Saf 2013;12: 187–93. 49. Perez-Sola MJ, Torre-Cisneros J, Perez-Zafrilla B, Carmona L, Descalzo MA, Gomez-Reino JJ. Infections in patients treated with tumor necrosis factor antagonists: incidence, etiology and mortality in the BIOBADASER registry. Med Clin (Barc) 2011; 137:533–40. 50. Rosenblum H, Amital H. Anti-TNF therapy: safety aspects of taking the risk. Autoimmun Rev 2011;10:563–8. 51. Rubbert-Roth A. Assessing the safety of biologic agents in patients with rheumatoid arthritis. Rheumatology (Oxford) 2012; 51(Suppl 5):v38–47. 52. Ruderman EM. Overview of safety of non-biologic and biologic DMARDs. Rheumatology (Oxford) 2012;51(Suppl 6):vi37–43. 53. Spinelli FR, Valesini G. Immunogenicity of anti-tumour necrosis factor drugs in rheumatic diseases. Clin Exp Rheumatol 2013;31: 954–63. 54. Tanaka Y, Hirata S, Saleem B, Emery P. Discontinuation of biologics in patients with rheumatoid arthritis. Clin Exp Rheumatol 2013;31(4 Suppl 78):S22–7. 55. Zhou H, Jang H, Fleischmann RM, et al. Pharmacokinetics and safety of golimumab, a fully human anti-TNF-alpha monoclonal antibody, in subjects with rheumatoid arthritis. J Clin Pharmacol 2007;47:383–96. 56. Marotte H, Charrin JE, Miossec P. Infliximab-induced aseptic meningitis. Lancet 2001;358:1784. 57. van Oosten BW, Barkhof F, Truyen L, et al. Increased MRI activity and immune activation in two multiple sclerosis patients treated with the monoclonal anti-tumor necrosis factor antibody cA2. Neurology 1996;47:1531–4. 58. Ordas I, Mould DR, Feagan BG, Sandborn WJ. Anti-TNF monoclonal antibodies in inflammatory bowel disease: pharmacokinetics-based dosing paradigms. Clin Pharmacol Ther 2012;91:635–46. 59. Vincent FB, Morand EF, Murphy K, Mackay F, Mariette X, Marcelli C. Antidrug antibodies (ADAb) to tumour necrosis factor (TNF)-specific neutralising agents in chronic inflammatory diseases: a real issue, a clinical perspective. Ann Rheum Dis 2013; 72:165–78. 60. van Kuijk AW, de Groot M, Stapel SO, Dijkmans BA, Wolbink GJ, Tak PP. Relationship between the clinical response to adalimumab treatment and serum levels of adalimumab and anti-adalimumab antibodies in patients with psoriatic arthritis. Ann Rheum Dis 2010;69:624–5. 61. Wang SL, Hauenstein S, Ohrmund L, et al. Monitoring of adalimumab and antibodies-to-adalimumab levels in patient serum by the homogeneous mobility shift assay. J Pharm Biomed Anal 2013;78–79:39–44. 62. Ordas I, Feagan BG, Sandborn WJ. Therapeutic drug monitoring of tumor necrosis factor antagonists in inflammatory bowel disease. Clin Gastroenterol Hepatol 2012;10:1079–87. quiz e85–6. 63. Garces S, Demengeot J, Benito-Garcia E. The immunogenicity of anti-TNF therapy in immune-mediated inflammatory diseases: a systematic review of the literature with a meta-analysis. Ann Rheum Dis 2013;72:1947–55. 64. Nanda KS, Cheifetz AS, Moss AC. Impact of antibodies to infliximab on clinical outcomes and serum infliximab levels in patients with inflammatory bowel disease (IBD): a meta-analysis. Am J Gastroenterol 2013;108:40–7. quiz 8. 65. Felson DT, Smolen JS, Wells G, et al. American College of Rheumatology/European League Against Rheumatism provisional definition of remission in rheumatoid arthritis for clinical trials. Arthritis Rheum 2011;63:573–86.

REV 5.2.0 DTD  TRSL830_proof  7 October 2014  6:35 pm  ce

1241 1242 1243 1244 1245 1246 1247 1248 1249 1250 1251 1252 1253 1254 1255 1256 1257 1258 1259 1260 1261 1262 1263 1264 1265 1266 1267 1268 1269 1270 1271 1272 1273 1274 1275 1276 1277 1278 1279 1280 1281 1282 1283 1284 1285 1286 1287 1288 1289 1290 1291 1292 1293 1294

Translational Research Volume -, Number -

1295 1296 1297 1298 1299 1300 1301 1302 1303 1304 1305 1306 1307 1308 1309 1310 1311 1312 1313 1314 1315 1316 1317 1318 1319 1320 1321 1322 1323 1324 1325 1326 1327 1328 1329 1330 1331 1332 1333 1334 1335 1336 1337 1338 1339 1340 1341 1342 1343 1344 1345 1346 1347 1348

Willrich et al

66. Neovius M, Simard JF, Askling J. Nationwide prevalence of rheumatoid arthritis and penetration of disease-modifying drugs in Sweden. Ann Rheum Dis 2011;70:624–9. 67. Chatzidionysiou K, Askling J, Eriksson J, Kristensen LE, van Vollenhoven R. Effectiveness of TNF inhibitor switch in RA: results from the national Swedish register. Ann Rheum Dis 2014. 68. Gomez-Reino JJ, Rodriguez-Lozano C, Campos-Fernandez C, Montoro M, Descalzo MA, Carmona L. Change in the discontinuation pattern of tumour necrosis factor antagonists in rheumatoid arthritis over 10 years: data from the Spanish registry BIOBADASER 2.0. Ann Rheum Dis 2012;71:382–5. 69. Jamnitski A, Krieckaert CL, Nurmohamed MT, et al. Patients non-responding to etanercept obtain lower etanercept concentrations compared with responding patients. Ann Rheum Dis 2012; 71:88–91. 70. Daien CI, Daien V, Parussini E, Dupuy AM, Combe B, Morel J. Etanercept concentration in patients with rheumatoid arthritis and its potential influence on treatment decisions: a pilot study. J Rheumatol 2012;39:1533–8. 71. Maser EA, Villela R, Silverberg MS, Greenberg GR. Association of trough serum infliximab to clinical outcome after scheduled maintenance treatment for Crohn’s disease. Clin Gastroenterol Hepatol 2006;4:1248–54. 72. Colombel JF, Sandborn WJ, Allez M, et al. Association between plasma concentrations of certolizumab pegol and endoscopic outcomes of patients with Crohn’s disease. Clin Gastroenterol Hepatol 2014;12:423–431.e1. 73. Steenholdt C. Use of infliximab and anti-infliximab antibody measurements to evaluate and optimize efficacy and safety of infliximab maintenance therapy in Crohn’s disease. Dan Med J 2013;60: B4616. 74. Svenson M, Geborek P, Saxne T, Bendtzen K. Monitoring patients treated with anti-TNF-alpha biopharmaceuticals: assessing serum infliximab and anti-infliximab antibodies. Rheumatology (Oxford) 2007;46:1828–34. 75. Yarur AJ, Abreu MT, Deshpande AR, Kerman DH, Sussman DA. Therapeutic drug monitoring in patients with inflammatory bowel disease. World J Gastroenterol 2014;20:3475–84. 76. Afif W, Loftus EV Jr, Faubion WA, et al. Clinical utility of measuring infliximab and human anti-chimeric antibody concentrations in patients with inflammatory bowel disease. Am J Gastroenterol 2010;105:1133–9. 77. Bendtzen K, Geborek P, Svenson M, Larsson L, Kapetanovic MC, Saxne T. Individualized monitoring of drug bioavailability and

78.

79.

80.

81.

82.

83.

84.

85.

86.

87.

13

immunogenicity in rheumatoid arthritis patients treated with the tumor necrosis factor alpha inhibitor infliximab. Arthritis Rheum 2006;54:3782–9. Paul S, Del Tedesco E, Marotte H, et al. Therapeutic drug monitoring of infliximab and mucosal healing in inflammatory bowel disease: a prospective study. Inflamm Bowel Dis 2013;19: 2568–76. Steenholdt C, Al-khalaf M, Brynskov J, Bendtzen K, Thomsen OO, Ainsworth MA. Clinical implications of variations in anti-infliximab antibody levels in patients with inflammatory bowel disease. Inflamm Bowel Dis 2012;18:2209–17. Steenholdt C, Bendtzen K, Brynskov J, Thomsen OO, Ainsworth MA. Cut-off levels and diagnostic accuracy of infliximab trough levels and anti-infliximab antibodies in Crohn’s disease. Scand J Gastroenterol 2011;46:310–8. Vande Casteele N, Ballet V, Van Assche G, Rutgeerts P, Vermeire S, Gils A. Early serial trough and antidrug antibody level measurements predict clinical outcome of infliximab and adalimumab treatment. Gut 2012;61:321. author reply 322. Vande Casteele N, Breynaert C, Vermeire S, Rutgeerts PJ, Gils A, Van Assche G. Incidence of acute severe infusion reactions to infliximab depends on definition used rather than assay. Aliment Pharmacol Ther 2011;34:401–3. author reply 4–5. Ainsworth MA, Bendtzen K, Brynskov J. Tumor necrosis factoralpha binding capacity and anti-infliximab antibodies measured by fluid-phase radioimmunoassays as predictors of clinical efficacy of infliximab in Crohn’s disease. Am J Gastroenterol 2008; 103:944–8. Vande Casteele N, Buurman DJ, Sturkenboom MG, et al. Detection of infliximab levels and anti-infliximab antibodies: a comparison of three different assays. Aliment Pharmacol Ther 2012;36: 765–71. Lallemand C, Kavrochorianou N, Steenholdt C, et al. Reporter gene assay for the quantification of the activity and neutralizing antibody response to TNFalpha antagonists. J Immunol Methods 2011;373:229–39. Wang SL, Ohrmund L, Hauenstein S, et al. Development and validation of a homogeneous mobility shift assay for the measurement of infliximab and antibodies-to-infliximab levels in patient serum. J Immunol Methods 2012;382:177–88. Willrich MAV, Barnidge DR, Karras RM, Ladwig PM, Murray DL, Snyder MR. A163 Infliximab measurement in serum using selective reaction monitoring LC-MS/MS. Clin Chem 2013; 59:A46.

REV 5.2.0 DTD  TRSL830_proof  7 October 2014  6:35 pm  ce

1349 1350 1351 1352 1353 1354 1355 1356 1357 1358 1359 1360 1361 1362 1363 1364 1365 1366 1367 1368 1369 1370 1371 1372 1373 1374 1375 1376 1377 1378 1379 1380 1381 1382 1383 1384 1385 1386 1387 1388 1389 1390 1391 1392 1393 1394 1395 1396 1397 1398 1399 1400 1401 1402

Tumor necrosis factor inhibitors: clinical utility in autoimmune diseases.

Tumor necrosis factor (TNF) production is amplified in several autoimmune disorders. In the 1990s, it became a validated therapeutic target used for t...
2MB Sizes 0 Downloads 9 Views