throbber
T r a s t u z u m a b E m t a n s i n e ( T - D M 1 ) : H i t c h i n g a
`R i d e o n a T h e r a p e u t i c A n t i b o d y
`
`By Howard A. Burris III, MD
`
`The treatment of cancers with chemotherapy is
`Overview:
`frequently limited by side effects. The effectiveness may be
`improved by the use of monoclonal antibodies to deliver
`cytotoxic agents to cancer cells while limiting exposure to
`normal tissues. The use of antibody-drug conjugates (ADCs) is
`one such strategy: a drug connected by a linker to an antibody
`specific for a tumor antigen is the basic makeup of an ADC.
`Overexpression and amplification of HER2 is associated with
`clinically aggressive breast cancers, and the use of trastu-
`zumab to target HER2 has been highly effective. That said,
`most patients with HER2-positive metastatic breast cancer
`will eventually have disease progression during targeted ther-
`apy. Trastuzumab emtansine (T–DM1)
`is a novel ADC that
`combines the humanized antibody trastuzumab and the potent
`antimicrotubule agent T-DM1 (derivative of maytansine) using
`a unique and highly stable linker. The potential of maytansine
`was found in the 1970s with clinical responses noted against
`breast cancer; however, substantial toxicity prohibited further
`development. DM1 possesses in vitro cytotoxicity 10 to 200
`times greater than that of taxanes and vinca alkaloids. A phase
`I trial of T-DM1 for patients with heavily pretreated HER2-
`positive breast cancer determined a recommended dose of 3.6
`
`THE TREATMENT of many cancers is often based on the
`
`use of standard chemotherapy agents and regimens.
`The effectiveness of this approach is frequently limited by
`substantial systemic toxicities associated with these agents.
`The therapeutic index can be markedly improved with the
`use of monoclonal antibodies to deliver potent cytotoxic
`agents to cancer cells, thus minimizing the exposure of the
`agents to normal tissues. The use of antibody-drug conju-
`gates (ADCs) is one such strategy; a cytotoxic drug con-
`nected by a chemical linker to a monoclonal antibody specific
`for a tumor antigen is the basic makeup of an ADC. More
`stable linkers and different cytotoxic agents have enabled a
`new generation of ADCs to enter the clinic. First and
`foremost, the design of an ADC centers on the selection of an
`antigen that is tumor-specific and accessible to antibody
`binding at the tumor cell.
`Overexpression and amplification of HER2 is associated
`with clinically aggressive breast cancers that have histori-
`cally had an overall poor prognosis and therapeutic resis-
`tance to traditional drugs. The use of trastuzumab to target
`the extracellular domain of HER2 has been highly effective
`in the treatment of this type of breast cancer. Multiple
`mechanisms for the efficacy of trastuzumab have been
`proposed, including inhibition of the PI3K signal transduc-
`tion pathway, antibody-dependent cell-mediated cytotoxic-
`ity (ADCC), and induction of apoptosis. When combined with
`chemotherapy, trastuzumab improves the time to disease
`progression and overall survival for patients with HER2-
`positive metastatic breast cancer. Furthermore, mature
`data from four large phase III trials in which trastuzumab
`was evaluated in the adjuvant setting, have demonstrated
`marked improvements in both disease-free and overall sur-
`vival. That said, most patients with HER2-positive meta-
`static breast cancer will eventually have disease progression
`during targeted therapy while the cancer continues to both
`
`mg per kilogram delivered every 3 weeks. Responses were
`seen in multiple patients. T-DM1 was then studied in phase II
`trials of patients with HER2-positive metastatic breast cancer.
`In a studies of 112 and 110 patients in whom disease had
`progressed during HER2-directed therapy, T-DM1 was asso-
`ciated with objective response rates of 26% and 34%, respec-
`tively. The agent was well tolerated in both trials, with most
`toxicities being grade 1 and 2, and no bleeding episodes or
`cardiac events occurring. Additional phase II and III trials are
`now evaluating T-DM1 in the first-line setting.
`In one such
`trial, T-DM1 was compared with standard dosing of trastu-
`zumab every 3 weeks plus docetaxel every 3 weeks. Objective
`response rates were comparable and grade 3 or4 adverse
`events were substantially reduced in the T-DM1 arm. The
`anticipated selective activity and reduction in side effects
`were thus noted. Randomized multicenter phase III trials are
`ongoing,
`including the EMILIA trial, an open-label
`trial of
`T-DM1 compared with the U.S. Food & Drug Administration-
`approved regimen of capecitabine plus lapatinib. The results
`of studies completed to date suggest T-DM1 is active in
`patients who have cancer resistant
`to trastuzumab-based
`combinations.
`
`express HER2 and demonstrate sensitivity to antimicrotu-
`bule agents.
`T-DM1 is a novel ADC that combines the humanized
`antibody trastuzumab and the potent antimicrotubule agent
`DM1 (derivative of maytansine) using a unique and highly
`stable linker.1 T-DM1, with its ability to bind HER2 with the
`same affinity as trastuzumab, maintains the activity of
`trastuzumab in addition to providing intracellular delivery
`of the antimicrotubule agent DM1. It is hypothesized that
`when T-DM1 binds to HER2 receptors, a portion of them
`undergo receptor internalization, followed by lysosomal deg-
`radation. Activated DM1 is then released from lysosome into
`the cellular cytoplasm after antibody degradation, inhibiting
`microtubule assembly and causing cell death. Potent cyto-
`toxic agents are needed to maximize the role of drug conju-
`gates. In addition, the drug must be inactive and nontoxic in
`the conjugated form to avoid systemic toxicities. Few agents
`are able to fulfill these characteristics, including the inhib-
`itors of tubulin polymerization (the maytansinoids and the
`auristatins).
`The potential of maytansine as an anticancer agent was
`originally discovered in the 1970s with clinical responses
`noted against breast cancer. However, substantial and ran-
`dom toxicities of neuropathy and myelosuppression were
`prohibitive of further clinical development. Recently, an
`attempt at improving the therapeutic index through conju-
`gation with trastuzumab was undertaken, leading to the
`
`From the Sarah Cannon Research Institute, Nashville, TN.
`Author’s disclosures of potential conflicts of interest are found at the end of this article.
`Address reprint requests to Howard A. Burris, MD, Tennessee Oncology/Sarah Cannon
`Research Institute, 250 25th Avenue N., Suite 110, Nashville, TN 37203; email:
`Howard.burris@scresearch.net
`© 2012 by American Society of Clinical Oncology.
`1092-9118/10/1-10
`
`159
`
`IMMUNOGEN 2188, pg. 1
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Table 1. Summary of Clinical Efficacy Data from Trastuzumab Emtasine (T-DM1) Clinical Trials
`
`HOWARD A. BURRIS III
`
`Trial and Reference
`
`TDM3569g2
`
`TDM4258g3
`
`TDM4374g4
`
`TDM4373g5
`
`TDM4450g6
`
`Treatment Regimens
`
`T-DM1 0.3 tp 4.8 mg/kg q3w for previously treated HER2⫹
`MBC after previous chemotherapy and disease progression
`on trastuzumab
`T-DM1 3.6 mg/kg q3w for HER2-positive MBC after previous
`chemotherapy and disease progression on HER2-targeted
`therapy
`T-DM1 3.6 mg/kg q3w for HER2-positive MBC after previous
`exposure to an anthracycline, a taxane, capecitabine and 2
`HER2-directed therapies in the metastatic setting
`T-DM1 3.6 mg/kg q3w ⫹ pertuzumab 840 mg loading dose
`then 420 mg q3w, for HER2⫹ MBC first-line treatment or
`after previous chemotherapy and HER2-directed therapy
`T-DM1 3.6 mg/kg q3w
`Or
`Trastuzumab 8 mg/kg loading dose then 6 mg/kg q3w ⫹
`docetaxel 75 mg/m2 or 100 mg/m2 q3w
`
`Number of
`Patients
`
`24
`
`112
`
`110
`
`67
`
`67
`70
`
`ORR, Number of
`Patients (%)
`
`6/24 (25.0)
`
`29 (26)
`
`38 (34.5)
`
`First line: 9/22 (41),
`Relapsed: 19/45 (42)
`
`32 (47.8)
`29 (41.4)
`
`Abbreviations: MBC, metastatic breast cancer; MTD, maximum tolerated dose; NR, not reported; ORR, objective response rate; q3w, every 3 weeks.
`
`development of DM1 (derivative of maytansine 1). DM1
`possesses in vitro cytotoxicity that is 10 to 200 times greater
`than that of other tubulin inhibitors, such as taxanes and
`vinca alkaloids. A suitable linker is critical to this process,
`and it must have a higher degree of stability in the circula-
`tion and allow efficient release of the potent cytotoxic agent
`once inside the tumor cell. The cytotoxic drug DM1 is
`conjugated to lysine residues of trastuzumab using a unique
`hetero-bifunctional
`reagent,
`N-succinimidyl
`4-(N-
`maleimidomethyl) cyclohexane-1-carboxylate (SMCC), in a
`two-step process. Trastuzumab is initially reacted with
`SMCC to form trastuzuamb-MCC. Next, T-MCC is then
`conjugated to DM1 to make T-DM1. The thioether linker
`was developed to provide a bond between trastuzumab and
`
`KEY POINTS
`● The therapeutic index for treating patients can be
`improved by the use of monoclonal antibodies to
`deliver potent cytotoxic agents to cancer cells while
`minimizing exposure of the agents to normal tissues.
`● The use of antibody-drug conjugates (ADCs) is one
`such strategy; a cytotoxic drug connected by a chem-
`ical linker to a monoclonal antibody specific for a
`tumor antigen is the basic makeup of an ADC.
`● Trastuzumab emtansine (T–DM1) is a novel ADC
`that combines the humanized antibody trastuzumab
`and the potent antimicrotubule agent T-DM1 (deriv-
`ative of maytansine) with use of a unique and highly
`stable linker.
`● In phase II studies of patients in whom metastatic
`breast cancer previously progressed during multiple
`HER2-directed therapies, T-DM1 was associated
`with response rates of 26% to 34% and was reason-
`ably well tolerated.
`● The results of phase II studies suggest T-DM1 can
`improve outcomes for patients with cancers that are
`resistant to trastuzumab-based combinations, and
`phase III trials are underway.
`
`160
`
`DM1 that is more stable than hydrazone or disulfide linkers.
`The therapeutic index of DM1 is thus enhanced by minimiz-
`ing systemic exposure to free DM1 and improving exposure
`to T-DM1. Before development in the clinic, the conjugate
`was extensively studied in preclinical models. The effective-
`ness of T-DM1 was established in three murine models of
`HER2-expressing human breast carcinoma. In contrast,
`little activity was seen in the normal human cells or breast
`cancer cells not overexpressing HER2, demonstrating the
`specificity of the ADC.1
`T-DM1 was the first HER2-targeted ADC with this unique
`SMCC linker to be studied in patients. A phase I trial in
`patients with HER2-positive breast cancer that had pro-
`gressed during prior trastuzumab-based therapy deter-
`mined a maximum tolerated dose of 3.6 mg per kilogram,
`delivered every 3 weeks.2 The dose-limiting toxicity was
`grade 4 thrombocytopenia that was rapidly reversible and
`not associated with clinically meaningful bleeding events.
`No cardiac events or left ventricular ejection fraction de-
`clines were noted. In addition, no alopecia greater than
`grade 1 was noted, further evidence for the lack of systemic
`toxicity.
`Six of the 24 patients had an objective partial response.
`All patients had previously been treated with trastuzumab,
`with a median exposure of approximately 2 years, as well as
`microtubulin inhibiting agents. Of the six responses, four
`occurred in the nine patients treated at the maximum
`tolerated dose. The trial pharmacokinetics demonstrated
`peak free (unconjugated) DM1 plasma concentrations imme-
`diately after dosing which were low on all time points,
`suggesting that any systemic toxicity was unrelated to
`circulating unconjugated DM1. Weekly dosing was also
`explored and was both active and well tolerated, but it
`showed no particular advantage from either a dose intensity
`or density standpoint.
`After these results, T-DM1 was studied in phase II trials
`of patients with HER2-positive metastatic breast cancer at
`the recommended dose of 3.6 mg per kilogram every 3 weeks.
`In a study of 112 patients who had disease progression
`during HER2-directed therapy, T-DM1 was associated with
`an objective response rate of 26% based on independent
`review, and progression-free survival of 4.6 months.3 The
`
`IMMUNOGEN 2188, pg. 2
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`T-DM1 AND THERAPEUTIC ANTIBODIES
`
`agent was well tolerated, with most toxicities being grade 1
`and 2, and no bleeding episodes or cardiac events.
`A second trial was conducted in 110 patients with HER2-
`positive metastatic breast cancer who had received prior
`treatment with an anthracycline, a taxane, capecitabine,
`trastuzumab, and lapatinib (and had had disease progres-
`sion during treatment with the most recent regimen).4
`T-DM1 demonstrated an objective response rate of 34.5%
`and a median progression-free survival of 6.9 months,
`based on independent reviews. The agent was again well
`tolerated in this heavily pretreated population, and no
`cardiac toxicity signals were noted. During these phase II
`trials, central review for HER2-positivity was required.
`Response rates were higher amongpatients with centrally
`verified HER2-positive tumors, whereas few responses were
`noted among patients with tumors that tested negatively on
`central review, confirming the relationship with drug activ-
`ity.
`Additional trials are now being done to evaluat T-DM1
`asfirst-line treatment. One such trial involved 137 patients
`who received either T-DM1 or standard dosing of trastu-
`zumab every 3 weeks plus docetaxel 75 or 100 mg/m2 every
`3 weeks.5 Objective response rates were comparable, at 48%
`and 41%, respectively. Of note, grade 3 or 4 adverse events
`were substantially reduced in the T-DM1 arm (37% com-
`pared with 75%). The selective activity and proposed reduc-
`tion in side effects were demonstrated in this randomized
`phase II trial.
`Two randomized multicenter phase III trials are ongoing
`to evaluate the role of T-DM1 in earlier lines of therapy.
`MARIANNE is designed to compare the efficacy and safety
`of single-agent T-DM1, alone or in combination with pertu-
`
`Author’s Disclosures of Potential Conflicts of Interest
`
`zumab, with the standard trastuzumab plus a taxane (pac-
`litaxel or docetaxel). The exposure to pertuzumab is blinded,
`and the standard arm is open-label.6
`Another study, EMILIA, is an open-label trial in which
`T-DM1 is being compared with the U.S. Food and Drug
`Administration (FDA)-approved regimen of capecitabine
`plus lapatinib in patients previously treated with both a
`taxane and trastuzumab. Interestingly, in another trial,
`T-DM1 is being compared with physician’s choice of treat-
`ment in patients who had disease progression after multiple
`prior regimens. Trastuzumab alone or with chemotherapy is
`allowed in the physician’s-choice arm.
`
`Conclusion
`
`The discovery of HER2 gene amplification and the subse-
`quent development of trastuzumab has markedly improved
`the prognosis for patients with HER2-positive breast cancer.
`Unfortunately, not all patients have a response to trastu-
`zumab, and disease will progress in most patients with
`metastatic HER2-positive disease. T-DM1 meets the criteria
`for a successful ADC by combining the targeted effect of
`trastuzumab with the cytotoxic potency of DM1 using a
`stable linker and minimizing systemic toxicity. In addition,
`other tumor histologies, such as gastrointestinal cancers
`that are HER2-positive may be sensitive to this agent. The
`results of phase II studies suggest T-DM1 can improve
`outcomes for patients with cancers that are resistant to
`trastuzumab-based combinations. An aggressive portfolio of
`phase II and III clinical trials will help determine the role
`of T-DM1 in earlier lines of therapy or with combinations of
`other targeted agents.
`
`Employment or
`Leadership
`Positions
`
`Author
`Howard A. Burris III*
`
`*No relevant relationships to disclose.
`
`Consultant or
`Advisory Role
`
`Stock
`Ownership
`
`Honoraria
`
`Research
`Funding
`
`Expert
`Testimony
`
`Other
`Remuneration
`
`REFERENCES
`1. Burris HA. Trastuzumab emtansine: A novel antibody-drug conjugate
`anthracycline, a taxane, capecitabine, lapatinib, and trastuzumab. Paper
`for HER2-positive breast cancer. Expert Opin Bio Ther. 2011;11:807-819.
`presented at: the European Society for Medical Oncology. October 10, 2010,
`2. Krop IE, Beeram M, Modi S, et al. Phase I study of trastuzumab-
`Milan, Italy, abstract 277O.
`DM1, a HER2 antibody-drug conjugate, given every 3 weeks to patients
`5. Perez EA, Dirix L, Kocsis J, et al. Efficacy and safety of trastuzumab-
`with HER2-positive metastatic breast cancer. J Clin Oncol. 2010;28:2698-
`DM1 versus trastuzumab plus docetaxel in HER2-positive metastatic breast
`2704.
`cancer patients with no prior chemotherapy for metastatic disease: Prelimi-
`3. Burris HA 3rd, Rugo HS, Vukelja SJ, et al. Phase II study of the antibody
`nary results of a randomized, multicenter open-label phase 2 (TDM4450g).
`drug conjugate trastuzumab-DM1 for the treatment of human epidermal
`Ann Oncol. 2010; 21 Suppl 8:LBA3.
`growth factor receptor 2 (HER2)-positive breast cancer after prior HER2-
`6. A study of trastuzumab-MCC-DM1 (T-DM1) in combination with pertu-
`directed therapy. J Clin Oncol. 2011;29:398-405.
`zumab administered to patients with HER2-positive locally advanced or
`4. Krop I, Lorusso P, Miller KD, et al. A phase II study of trastuzumab-
`metastatic breast cancer who have previously received trastuzumab. http://
`www.clinicaltrials.gov/ct2/show/NCT00875979?term⫽t-dm1⫹4373&rank⫽1.
`DM1 (T-DM1), a novel HER2 antibody-drug conjugate, in patients with
`HER2-positive metastatic breast cancer who were previously treated with an
`Accessed August 3, 2010.
`
`161
`
`IMMUNOGEN 2188, pg. 3
`Phigenix v. Immunogen
`IPR2014-00676
`
`

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket